1
|
Ostertag F, Grimm VJ, Hinrichs J. Iron saturation and binding capacity of lactoferrin - development and validation of a colorimetric protocol for quality control. Food Chem 2024; 463:141365. [PMID: 39332363 DOI: 10.1016/j.foodchem.2024.141365] [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: 07/10/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Among the numerous biofunctional properties of lactoferrin, its ability to bind iron ions can be considered a core function. The saturation level with ferric iron affects the stability and functionality of the protein. To reliably quantify the iron saturation, an assay based on the color reagent Ferrozine was developed and validated concerning the lower detection (0.023 μg mL-1) and quantification limits (0.069 μg mL-1), as well as precision, recovery and accuracy values. The established assay was used to monitor iron uptake, comparing two commercially available bovine lactoferrin powders. Significant differences between the samples were observed. One sample exhibited nearly ideal binding behavior with a high affinity for ferric iron (saturation > 98 %), while the comparison sample did not exceed saturation values >80 %. This finding underscores the importance of assessing the iron status and binding capacity for the quality evaluation of lactoferrin products.
Collapse
Affiliation(s)
- Fabian Ostertag
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany.
| | - Vanessa J Grimm
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany
| | - Jörg Hinrichs
- University of Hohenheim, Institute of Food Science and Biotechnology, Department of Soft Matter Science and Dairy Technology, Garbenstrasse 21, 70599 Stuttgart, Germany
| |
Collapse
|
2
|
Tiwari R, Tickell KD, Yoshioka E, Otieno J, Shah A, Richardson BA, Keter L, Okello M, Nyabinda C, Trehan I, McGrath CJ, Means AR, Houpt ER, Liu J, Platts-Mills JA, Njunge JM, Rwigi D, Diakhate MM, Nyaoke J, Ochola E, John-Stewart G, Walson JL, Pavlinac PB, Singa BO. Lactoferrin and lysozyme to promote nutritional, clinical and enteric recovery: a protocol for a factorial, blinded, placebo-controlled randomised trial among children with diarrhoea and malnutrition (the Boresha Afya trial). BMJ Open 2024; 14:e079448. [PMID: 39122384 PMCID: PMC11331881 DOI: 10.1136/bmjopen-2023-079448] [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: 08/31/2023] [Accepted: 06/21/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Children with moderate or severe wasting are at particularly high risk of recurrent or persistent diarrhoea, nutritional deterioration and death following a diarrhoeal episode. Lactoferrin and lysozyme are nutritional supplements that may reduce the risk of recurrent diarrhoeal episodes and accelerate nutritional recovery by treating or preventing underlying enteric infections and/or improving enteric function. METHODS AND ANALYSIS In this factorial, blinded, placebo-controlled randomised trial, we aim to determine the efficacy of lactoferrin and lysozyme supplementation in decreasing diarrhoea incidence and improving nutritional recovery in Kenyan children convalescing from comorbid diarrhoea and wasting. Six hundred children aged 6-24 months with mid-upper arm circumference <12.5 cm who are returning home after an outpatient visit or inpatient hospital stay for diarrhoea will be enrolled. Children will be randomised to 16 weeks of lactoferrin, lysozyme, a combination of the two, or placebo and followed for 24 weeks, with biweekly home visits by community health workers and clinic visits at 4, 10, 16 and 24 weeks. The primary analysis will compare the incidence of moderate-to-severe diarrhoea and time to nutritional recovery between each intervention arm and placebo. The trial will also test whether these interventions reduce enteric pathogen carriage, decrease enteric permeability and/or increase haemoglobin concentration in enrolled children. Finally, we will evaluate the acceptability, adherence and cost-effectiveness of lactoferrin and/or lysozyme. ETHICS AND DISSEMINATION The trial has been approved by the institutional review boards of the Kenya Medical Research Institute, the University of Washington, the Kenyan Pharmacy and Poisons Board, and the Kenyan National Commission on Science, Technology and Innovation. The results of this trial will be shared with local and international stakeholders and published in peer-reviewed journals, and the key findings will be presented at relevant conferences. TRIAL REGISTRATION NUMBER NCT05519254, PACTR202108480098476.
Collapse
Affiliation(s)
- Ruchi Tiwari
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Kirkby D Tickell
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Emily Yoshioka
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Joyce Otieno
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Barbra A Richardson
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Lucia Keter
- Kenya Medical Research Institute, Nairobi, Nairobi County, Kenya
| | - Maureen Okello
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | | | - Indi Trehan
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Christine J McGrath
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Arianna Rubin Means
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Eric R Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, North Carolina, USA
| | - Jie Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, North Carolina, USA
| | - James M Njunge
- Centre for Geographic Medicine Research Coast, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Doreen Rwigi
- Kenya Medical Research Institute, Nairobi, Nairobi County, Kenya
- Kenya Medical Research Institute, Nairobi, Kenya
| | - Mareme M Diakhate
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Julius Nyaoke
- Kenya Medical Research Institute, Nairobi, Nairobi County, Kenya
| | - Eric Ochola
- Kenya Medical Research Institute, Nairobi, Nairobi County, Kenya
| | - Grace John-Stewart
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Judd L Walson
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Patricia B Pavlinac
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Benson O Singa
- Centre for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| |
Collapse
|
3
|
Rosa L, Ianiro G, Niro A, Musci G, Paesano R, Cutone A, Valenti P. Valpalf ®: A New Nutraceutical Formulation Containing Bovine Lactoferrin That Exhibits Potentiated Biological Activity. Int J Mol Sci 2024; 25:8559. [PMID: 39201246 PMCID: PMC11354537 DOI: 10.3390/ijms25168559] [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: 07/16/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
As a nutraceutical, bovine lactoferrin (bLf), an iron-binding glycoprotein involved in innate immunity, is gaining elevated attention for its ability to exert pleiotropic functions and to be exceptionally tolerated even at high dosages. Some of bLf's activities, including its anti-inflammatory and antioxidant, are tightly linked to its ability to both chelate iron and enter inside the cell nucleus. Here, we present data about Valpalf®, a new formulation containing bLf, sodium citrate, and sodium bicarbonate at a molar ratio of 10-3. In the present study, Valpalf® exhibits superior iron-binding capacity, resistance to tryptic digestion, and a greater capacity to accumulate into the nucleus over time when compared to the native bLf alone. In agreement, Valpalf® effectively reduces interleukin(IL)-6 levels in lipopolysaccharide-stimulated macrophages and modulates the expression of antioxidant enzymes, such as superoxide dismutase 1 and 2, in phorbol-12-myristate-13-acetate-stimulated monocytes. Of note, this potentiated bioactivity was corroborated in a retrospective study on the treatment of anemia of inflammation in hereditary thrombophilic pregnant and non-pregnant women, demonstrating that Valpalf® improves hematological parameters and reduces serum IL-6 levels to a higher extent than bLf alone.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antonella Niro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | | | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy; (L.R.); (P.V.)
- Microbo s.r.l., 00153 Rome, Italy;
| |
Collapse
|
4
|
Gu H, Wang Y, Wang Y, Ding L, Huan W, Yang Y, Fang F, Cui W. Global Bibliometric and Visualized Analysis of Research on Lactoferrin from 1978 to 2024. Mol Nutr Food Res 2024; 68:e2400379. [PMID: 39044343 DOI: 10.1002/mnfr.202400379] [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: 05/20/2024] [Revised: 07/02/2024] [Indexed: 07/25/2024]
Abstract
SCOPE Lactoferrin (LF) is an iron-bound protein with a molecular weight of about 80 kDa. LF has many biological functions such as antibacterial, antiviral, immunomodulatory, and anticancer. The purpose of this study is to explore the research trend of LF through bibliometric analysis. METHODS AND RESULTS The search is conducted in the Web of Science Core Collection database, and then the publications information of LF related literature is exported. Based on CiteSpace and VOSviewer software, countries, institutions, authors, journals, keywords, and so on are analyzed. Since 1987, a total of 9382 literature have been included, and the number of papers related to LF has increased year by year. These publications come mainly from 124 countries and 725 institutions. Of the 1256 authors analyzed, Valenti Piera is the one with the most publications. The burst strength of gut microbiota, antioxidant, nanoparticles, and in vitro digestion are 21.3, 15.63, 23.03, and 13.51, respectively. They represent the frontier of research in this field and are developing rapidly. CONCLUSION This study shows that LF has important research value. The study of LF nanoparticles and the effects of LF on the gut microbiota are an emerging field that helps to explore new research directions.
Collapse
Affiliation(s)
- Hong Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yiming Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yating Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Liyi Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Wenru Huan
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuting Yang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Fang Fang
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| | - Weiwei Cui
- Department of Nutrition and Food Hygiene, School of Public Health, Jilin University, Changchun, 130021, China
| |
Collapse
|
5
|
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
|
6
|
Wang W, An Q, Huang K, Dai Y, Meng Q, Zhang Y. Unlocking the power of Lactoferrin: Exploring its role in early life and its preventive potential for adult chronic diseases. Food Res Int 2024; 182:114143. [PMID: 38519174 DOI: 10.1016/j.foodres.2024.114143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 03/24/2024]
Abstract
Nutrition during the early postnatal period exerts a profound impact on both infant development and later-life health. Breast milk, which contains lactoferrin, a dynamic protein, plays a crucial role in the growth of various biological systems and in preventing numerous chronic diseases. Based on the relationship between early infant development and chronic diseases later in life, this paper presents a review of the effects of lactoferrin in early life on neonates intestinal tract, immune system, nervous system, adipocyte development, and early intestinal microflora establishment, as well as the preventive and potential mechanisms of early postnatal lactoferrin against adult allergy, inflammatory bowel disease, depression, cancer, and obesity. Furthermore, we summarized the application status of lactoferrin in the early postnatal period and suggested directions for future research.
Collapse
Affiliation(s)
- Wenli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qin An
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yunping Dai
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Qingyong Meng
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
| |
Collapse
|
7
|
Zhang Z, Zuo L, Song X, Wang L, Zhang Y, Cheng Y, Huang J, Zhao T, Yang Z, Zhang H, Li J, Zhang X, Geng Z, Wang Y, Ge S, Hu J. Arjunolic acid protects the intestinal epithelial barrier, ameliorating Crohn's disease-like colitis by restoring gut microbiota composition and inactivating TLR4 signalling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155223. [PMID: 38134862 DOI: 10.1016/j.phymed.2023.155223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 09/15/2023] [Accepted: 11/14/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND AIMS Crohn's disease (CD) is characterized by an overabundance of epithelial cell death and an imbalance in microflora, both of which contribute to the dysfunction of the intestinal barrier. Arjunolic acid (AA) has anti-apoptotic effects and regulates microbiota efficacy. The objective of this study was to assess the impact of the treatment on colitis resembling Crohn's disease, along with exploring the potential underlying mechanism. METHODS CD animal models were created using Il-10-/- mice, and the impact of AA on colitis in mice was evaluated through disease activity index, weight fluctuations, pathological examination, and assessment of intestinal barrier function. To clarify the direct role of AA on intestinal epithelial cell apoptosis, organoids were induced by LPS, and TUNEL staining was performed. To investigate the potential mechanisms of AA in protecting the intestinal barrier, various methods including bioinformatics analysis and FMT experiments were employed. RESULTS The treatment for AA enhanced the condition of colitis and the function of the intestinal barrier in Il-10-/- mice. This was demonstrated by the amelioration of weight loss, reduction in tissue inflammation score, and improvement in intestinal permeability. Moreover, AA suppressed the apoptosis of intestinal epithelial cells in Il-10-/- mice and LPS-induced colon organoids, while also reducing the levels of Bax and C-caspase-3. In terms of mechanism, AA suppressed the activation of TLR4 signaling in Il-10-/- mice and colon organoids induced by LPS. In addition, AA increased the abundance of short-chain fatty acid-producing bacteria in the stool of Il-10-/- mice, and transplantation of feces from AA-treated mice improved CD-like colitis. CONCLUSIONS The results of our study demonstrate that AA has a protective effect on the intestinal barrier in Crohn's disease-like colitis by preventing apoptosis. Additionally, this groundbreaking study reveals the capacity of AA to hinder TLR4 signaling and alter the makeup of the intestinal microbiome. The findings present fresh possibilities for treating individuals diagnosed with Crohn's disease. AA offers a hopeful novel strategy for managing Crohn's disease by obstructing crucial pathways implicated in intestinal inflammation and enhancing the gut microbiota.
Collapse
Affiliation(s)
- Zining Zhang
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China
| | - Xue Song
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Yan Zhang
- Bengbu Medical College, Bengbu, Anhui, China
| | - Yang Cheng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ju Huang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Tianhao Zhao
- Department of Gastroenterology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Jing Li
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu, Medical College, Bengbu, China
| | - Xiaofeng Zhang
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhijun Geng
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yueyue Wang
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu, Medical College, Bengbu, China
| | - Sitang Ge
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China
| | - Jianguo Hu
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical College, Bengbu, China; Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Anhui, China; Department of Clinical Laboratory, First Affiliated Hospital of Bengbu, Medical College, Bengbu, China.
| |
Collapse
|
8
|
Rodríguez-Camejo C, Puyol A, Arbildi P, Sóñora C, Fazio L, Siré G, Hernández A. Effects of human donor milk on gut barrier function and inflammation: in vitro study of the beneficial properties to the newborn. Front Immunol 2023; 14:1282144. [PMID: 38022652 PMCID: PMC10663376 DOI: 10.3389/fimmu.2023.1282144] [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: 08/23/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction The gastrointestinal and immune systems of premature infants are not fully developed, rendering them more vulnerable to severe complications like necrotizing enterocolitis. Human milk offers a rich array of bioactive factors that collectively contribute to reducing the incidence of gut infections and inflammatory conditions. When a mother's milk is unavailable, preterm infants are often provided with donor human milk processed in Human Milk Banks. However, it remains uncertain whether pasteurized milk confers the same level of risk reduction as unprocessed milk. This uncertainty may stem from the well-documented adverse effects of heat treatment on milk composition. Yet, our understanding of the comprehensive impact on protective mechanisms is limited. Methods In this study, we conducted a comparative analysis of the effects of raw versus pasteurized milk and colostrum versus mature milk on cellular functions associated with the gut epithelial barrier and responses to inflammatory stimuli. We utilized THP-1 and HT-29 cell lines, representing monocyte/macrophages and gut epithelial cells, respectively. Results Our observations revealed that all milk types stimulated epithelial cell proliferation. However, only raw colostrum increased cell migration and interfered with the interaction between E. coli and epithelial cells. Furthermore, the response of epithelial and macrophage cells to lipopolysaccharide (LPS) was enhanced solely by raw colostrum, with a milder effect observed with mature milk. In contrast, both raw and pasteurized milk diminished the LPS induced response in monocytes. Lastly, we examined how milk affected the differentiation of monocytes into macrophages, finding that milk reduced the subsequent inflammatory response of macrophages to LPS. Discussion Our study sheds light on the impact of human milk on certain mechanisms that potentially account for its protective effects against necrotizing enterocolitis, highlighting the detrimental influence of pasteurization on some of these mechanisms. Our findings emphasize the urgency of developing alternative pasteurization methods to better preserve milk properties. Moreover, identifying the key components critically affected by these protective mechanisms could enable their inclusion in donor milk or formula, thereby enhancing immunological benefits for vulnerable newborns.
Collapse
Affiliation(s)
- Claudio Rodríguez-Camejo
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
- Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Higiene “Prof. Arnoldo Berta”, Universidad de la República, Montevideo, Uruguay
| | - Arturo Puyol
- Banco de Leche “Ruben Panizza”, Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Paula Arbildi
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
- Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Higiene “Prof. Arnoldo Berta”, Universidad de la República, Montevideo, Uruguay
| | - Cecilia Sóñora
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
- Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Higiene “Prof. Arnoldo Berta”, Universidad de la República, Montevideo, Uruguay
- Escuela Universitaria de Tecnología Médica (EUTM), Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Laura Fazio
- Banco de Leche “Ruben Panizza”, Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Gabriela Siré
- Banco de Leche “Ruben Panizza”, Centro Hospitalario Pereira Rossell, Administración de los Servicios de Salud del Estado, Montevideo, Uruguay
| | - Ana Hernández
- Área Inmunología, Departamento de Biociencias (DEPBIO), Facultad de Química, Universidad de la República, Montevideo, Uruguay
- Unidad Asociada de Inmunología, Instituto de Química Biológica (IQB), Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
- Laboratorio de Inmunología, Instituto de Higiene “Prof. Arnoldo Berta”, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
9
|
Ostertag F, Hinrichs J. Enrichment of Lactoferrin and Immunoglobulin G from Acid Whey by Cross-Flow Filtration. Foods 2023; 12:foods12112163. [PMID: 37297408 DOI: 10.3390/foods12112163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
The production of cream cheese, curd, high-protein yogurt, or caseinate results in large amounts of acid whey as a by-product. So far acid whey is often disposed as animal feed or organic fertilizer. However, these approaches ignore the valorization potential that arises from the unique composition of the whey protein fraction. Whey contains the biofunctional proteins lactoferrin and immunoglobulin G, which possess immune-supporting, antibacterial, antiviral, and numerous further health-promoting functions. However, the concentration of these proteins in bovine milk or whey is below a physiologically relevant level. Based on literature research we specified a daily intake of 200 mg lactoferrin as the minimal functional dose. By means of cross-flow ultrafiltration, an attempt was made to increase the concentration of biofunctional proteins. Therefore, a membrane for the selective retention of lactoferrin and immunoglobulin G was identified, and the process parameters were optimized. Finally, a concentration experiment was conducted, whereby the concentration of biofunctional proteins was increased up to factor 30. The biofunctionality was assessed in a microbiological assay. Surprisingly, the antimicrobial growth inhibition of the produced concentrate was even higher than in pure lactoferrin. The presented approach offers a strategy to convert an abundant but underutilized by-product into valuable products for human nutrition.
Collapse
Affiliation(s)
- Fabian Ostertag
- Department of Soft Matter Science and Dairy Technology, Institute of Food Science and Biotechnology, University of Hohenheim, Garben Str. 21, 70599 Stuttgart, Germany
| | - Jörg Hinrichs
- Department of Soft Matter Science and Dairy Technology, Institute of Food Science and Biotechnology, University of Hohenheim, Garben Str. 21, 70599 Stuttgart, Germany
| |
Collapse
|
10
|
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: 9] [Impact Index Per Article: 9.0] [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
|
11
|
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: 3.0] [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
|
12
|
Sarkar VK, De UK, Kala A, Verma AK, Chauhan A, Paul BR, Soni S, Gandhar JS, Chaudhuri P, Patra MK, Eregowda CG, Gaur GK. Early-Life Intervention of Lactoferrin and Probiotic in Suckling Piglets: Effects on Immunoglobulins, Intestinal Integrity, and Neonatal Mortality. Probiotics Antimicrob Proteins 2023; 15:149-159. [PMID: 35793035 DOI: 10.1007/s12602-022-09964-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 01/18/2023]
Abstract
The aim of this study was to determine the effects of early-life bovine lactoferrin and host specific probiotic interventions on growth performance, mortality, and concentrations of immunoglobulin A and immunoglobulin G and transforming growth factor beta 1 (a marker of intestinal integrity) in serum of neonatal piglets. A total of eight piglet litters from parity matched sows were randomly divided into four groups and assigned to one of the four interventions: control (sterile normal saline), bovine lactoferrin (100 mg bovine lactoferrin), probiotic (1 × 109 colony forming unit (cfu) of swine origin Pediococcus acidilactici FT28 probiotic), and bovine lactoferrin + probiotic (100 mg bovine lactoferrin and 1 × 109 CFU of P. acidilactici FT28 probiotic). All the interventions were given once daily through oral route for first 7 days of life. The average daily gain (p = 0.0004) and weaning weight (p < 0.0001) were significantly improved in the probiotic group. The piglet survivability was significantly higher in bovine lactoferrin and probiotic groups than control group in Log-rank (Mantel-Cox) test. The concentrations of immunoglobulin A on day 21 in bovine lactoferrin, probiotic, and bovine lactoferrin + probiotic groups increased significantly (p < 0.05). Immunoglobulin G concentrations on day 7 and 15 in bovine lactoferrin and bovine lactoferrin + probiotic groups and on day 15 in probiotic group were significantly (p < 0.05) elevated, whereas, the concentration of transforming growth factor-β1 was significantly (p < 0.05) increased from day 7 to 21 in all the supplemented groups. In conclusion, the early-life bovine lactoferrin and P. acidilactici FT28 probiotic interventions reduced the mortality in the suckling piglets by promoting the systemic immunity and enhancing the intestinal integrity.
Collapse
Affiliation(s)
- Varun Kumar Sarkar
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Ujjwal Kumar De
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India.
| | - Anju Kala
- Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Ashok Kumar Verma
- Division of Animal Nutrition, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Anuj Chauhan
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Babul Rudra Paul
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Srishti Soni
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Jitendra Singh Gandhar
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Pallab Chaudhuri
- Division of Biological Products, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Manas Kumar Patra
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| | - Chethan Gollahalli Eregowda
- Department of Veterinary Medicine, College of Veterinary Sciences and Animal Husbandry, Central Agriculture University, Selesih, Aizawl, 796014, Mizoram, India
| | - Gyanendra Kumar Gaur
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, 243122 (UP), India
| |
Collapse
|
13
|
Guzmán-Mejía F, Godínez-Victoria M, Molotla-Torres DE, Drago-Serrano ME. Lactoferrin as a Component of Pharmaceutical Preparations: An Experimental Focus. Pharmaceuticals (Basel) 2023; 16:214. [PMID: 37259362 PMCID: PMC9961256 DOI: 10.3390/ph16020214] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 10/29/2023] Open
Abstract
Lactoferrin is an 80 kDa monomeric glycoprotein that exhibits multitask activities. Lactoferrin properties are of interest in the pharmaceutical field for the design of products with therapeutic potential, including nanoparticles and liposomes, among many others. In antimicrobial preparations, lactoferrin has been included either as a main bioactive component or as an enhancer of the activity and potency of first-line antibiotics. In some proposals based on nanoparticles, lactoferrin has been included in delivery systems to transport and protect drugs from enzymatic degradation in the intestine, favoring the bioavailability for the treatment of inflammatory bowel disease and colon cancer. Moreover, nanoparticles loaded with lactoferrin have been formulated as delivery systems to transport drugs for neurodegenerative diseases, which cannot cross the blood-brain barrier to enter the central nervous system. This manuscript is focused on pharmaceutical products either containing lactoferrin as the bioactive component or formulated with lactoferrin as the carrier considering its interaction with receptors expressed in tissues as targets of drugs delivered via parenteral or mucosal administration. We hope that this manuscript provides insights about the therapeutic possibilities of pharmaceutical Lf preparations with a sustainable approach that contributes to decreasing the resistance of antimicrobials and enhancing the bioavailability of first-line drugs for intestinal chronic inflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Fabiola Guzmán-Mejía
- Unidad Xochimilco, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Ciudad de México CP 04960, Mexico
| | - Marycarmen Godínez-Victoria
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México CP 11340, Mexico
| | - Daniel Efrain Molotla-Torres
- Unidad Xochimilco, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Ciudad de México CP 04960, Mexico
| | - Maria Elisa Drago-Serrano
- Unidad Xochimilco, Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana, Ciudad de México CP 04960, Mexico
| |
Collapse
|
14
|
Lactoferrin Restores the Deoxynivalenol-Impaired Spermatogenesis and Blood-Testis Barrier Integrity via Improving the Antioxidant Capacity and Modifying the Cell Adhesion and Inflammatory Response. Antioxidants (Basel) 2023; 12:antiox12010152. [PMID: 36671014 PMCID: PMC9855165 DOI: 10.3390/antiox12010152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/12/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Deoxynivalenol (DON) is among the most prevalent contaminants in cereal crops and has been demonstrated to impair male spermatogenesis and induce oxidative stress, testicular apoptosis, and disruption of the blood-testis barrier (BTB). Lactoferrin (LF) is an iron-binding glycoprotein with multifunctions including anti-inflammation and antioxidation. Thus, this study aimed to investigate the effects of LF on the spermatogenesis and integrity of the BTB in DON-exposed mice. Thirty-two male mice were allotted to four groups for a 35-day feeding period: vehicle (basal diet), DON (12 mg/kg), LF (10 mg/d, p.o.), and DON + LF. The results showed that DON induced vacuolization of the spermatogenic epithelium, broke the adhesion junction between Sertoli cells and spermatids established by N-cadherin and induced testicular oxidative stress. LF administration restored sperm production, attenuated the DON-induced oxidative stress and reduced the breakages in adhesion junction. DON exposure enhanced the protein expression of occludin. Transcriptional profiling of the testis observed a disturbance in the expression profiles of cell adhesion and inflammatory response genes, and LF administration reversed these gene expressions. Furthermore, down-regulated signaling pathways, including the apical junction, TNFα signaling via NF-κB, and TGF-β in the DON group were observed. These were restored by LF. Enrichment analysis between DON + LF group and vehicle also confirmed the absence of these pathways. These findings indicated that LF eliminated the DON-induced detriment to spermatogenesis and cell connections between Sertoli cells and spermatids via improving antioxidant capacity and modifying the inflammatory response and cell adhesion genes.
Collapse
|
15
|
Young G, Berrington JE, Cummings S, Dorling J, Ewer AK, Frau A, Lett L, Probert C, Juszczak E, Kirby J, Beck LC, Renwick VL, Lamb C, Lanyon CV, McGuire W, Stewart C, Embleton N. Mechanisms affecting the gut of preterm infants in enteral feeding trials: a nested cohort within a randomised controlled trial of lactoferrin. Arch Dis Child Fetal Neonatal Ed 2022; 108:272-279. [PMID: 36396443 PMCID: PMC10176413 DOI: 10.1136/archdischild-2022-324477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To determine the impact of supplemental bovine lactoferrin on the gut microbiome and metabolome of preterm infants. DESIGN Cohort study nested within a randomised controlled trial (RCT). Infants across different trial arms were matched on several clinical variables. Bacteria and metabolite compositions of longitudinal stool and urine samples were analysed to investigate the impact of lactoferrin supplementation. SETTING Thirteen UK hospitals participating in a RCT of lactoferrin. PATIENTS 479 infants born <32 weeks' gestation between June 2016 and September 2017. RESULTS 10 990 stool and 22 341 urine samples were collected. Analyses of gut microbiome (1304 stools, 201 infants), metabolites (171 stools, 83 infants; 225 urines, 90 infants) and volatile organic compounds (314 stools, 117 infants) were performed. Gut microbiome Shannon diversity at 34 weeks corrected age was not significantly different between infants in the lactoferrin (mean=1.24) or placebo (mean=1.06) groups (p=0.11). Lactoferrin receipt explained less than 1% variance in microbiome compositions between groups. Metabolomic analysis identified six discriminative features between trial groups. Hospital site (16%) and postnatal age (6%) explained the greatest variation in microbiome composition. CONCLUSIONS This multiomic study identified minimal impacts of lactoferrin but much larger impacts of hospital site and postnatal age. This may be due to the specific lactoferrin product used, but more likely supports the findings of the RCT in which this study was nested, which showed no impact of lactoferrin on reducing rates of sepsis. Multisite mechanistic studies nested within RCTs are feasible and help inform trial interpretation and future trial design.
Collapse
Affiliation(s)
- Greg Young
- Applied Sciences, Northumbria University Faculty of Health and Life Sciences, Newcastle upon Tyne, England, UK.,Microbial Environments, Hub for Biotechnology in the Built Environment, Newcastle upon Tyne, England, UK
| | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK .,Newcastle Neonatal Service, Ward 35 Neonatal Unit, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Stephen Cummings
- School of Health and Life Sciences, Teesside University, Middlesbrough, North Yorkshire, UK
| | - Jon Dorling
- Department of Neonatal Medicine, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew K Ewer
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Alessandra Frau
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Lauren Lett
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Chris Probert
- Gastroenterology Research Unit, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Ed Juszczak
- School of Medicine, University of Nottingham School of Medicine, Nottingham, Notts, UK
| | - John Kirby
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Lauren C Beck
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Victoria L Renwick
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Christopher Lamb
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Clare V Lanyon
- Applied Sciences, Northumbria University Faculty of Health and Life Sciences, Newcastle upon Tyne, England, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, North Yorkshire, UK
| | - Christopher Stewart
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Ward 35 Neonatal Unit, Royal Victoria Infirmary, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.,Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
16
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [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
|
17
|
Effects of dietary supplementation of bovine lactoferrin on growth performance, immune function and intestinal health in weaning piglets. Biometals 2022; 36:587-601. [PMID: 36342570 DOI: 10.1007/s10534-022-00461-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022]
Abstract
Weaning is a crucial period in the pig's life cycle, which is frequently followed by gastrointestinal (GI) infections, diarrhea and even death. This study focused on the impact of bovine lactoferrin (bLF) supplementation on the intestinal health of weaning piglets. Weaning piglets (Duroc × Landrace × Yorkshire, 23 days) were randomly allocated into four groups, which included negative control group (CON): basic diet; positive control group (ANT): basic diet + 20 mg/kg flavomycin + 100 mg/kg aureomycin; treatment group bLF-A: basic diet + 1 g/kg bLF; treatment group bLF-B: basic diet + 3 g/kg bLF. The result showed that dietary supplementation of bLF can improve growth performance and reduce diarrhea, which exhibits dose-dependency (P < 0.05). Compared with CON group, supplementation with bLF significantly improved immunity, and increased villus height and ratio of villus height/crypt depth at the small intestinal mucosa (P < 0.05). The mRNA expression of claudin-1, occludin and ZO-1 was greatly increased in the ileum of bLF group on days 7 and 14 (P < 0.05). Furthermore, the supplementation of bLF increased the abundance of Lactobacillus and Bifidobacterium and decreased the abundance of Escherichia coli in the cecum on day 7 (P < 0.05). The dietary supplementation of bLF enhanced the growth performance, reduced diarrhea rate in weaning piglets by improving intestinal immunity, morphology and barrier function, balancing intestinal microbiota. And bLF can be a promising feed additive in relieving stress situation of weaning piglets.
Collapse
|
18
|
Hu P, Zong Q, Zhao Y, Gu H, Liu Y, Gu F, Liu HY, Ahmed AA, Bao W, Cai D. Lactoferrin Attenuates Intestinal Barrier Dysfunction and Inflammation by Modulating the MAPK Pathway and Gut Microbes in Mice. J Nutr 2022; 152:2451-2460. [PMID: 36774111 DOI: 10.1093/jn/nxac200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/16/2022] [Accepted: 08/27/2022] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Deoxynivalenol (DON) is a major mycotoxin present in staple foods (particularly in cereal products) that induces intestinal inflammation and disrupts intestinal integrity. Lactoferrin (LF) is a multifunctional protein that contributes to maintaining intestinal homeostasis and improving host health. However, the protective effects of LF on DON-induced intestinal dysfunction remain unclear. OBJECTIVES This study aimed to investigate the effects of LF on DON-induced intestinal dysfunction in mice, and its underlying protective mechanism. METHODS Male BALB/c mice (5 wk old) with similar body weights were divided into 4 groups (n = 6/group) and treated as follows for 5 wk: Veh [peroral vehicle daily, commercial (C) diet]; LF (peroral 10 mg LF/d, C diet); DON (Veh, C diet containing 12 mg DON/kg); and LF + DON (peroral 10 mg LF/d, DON diet). Intestinal morphology, tight junction proteins, cytokines, and microbial community were determined. Data were analyzed by 2-factor ANOVA or Kruskal-Wallis test. RESULTS The DON group exhibited lower final body weight (-12%), jejunal villus height (VH; -41%), and jejunal occludin expression (-36%), and higher plasma IL-1β concentration (+85%) and jejunal Il1b mRNA expression (+98%) compared with the Veh group (P < 0.05). In contrast, final body weight (+19%), jejunal VH (+49%), jejunal occludin (+53%), and intelectin 1 protein expression (+159%) were greater in LF + DON compared with DON (P < 0.05). Additionally, jejunal Il1b mRNA expression (-31%) and phosphorylation of p38 and extracellular signal regulated kinase 1/2 (-40% and - 38%) were lower in LF + DON compared with DON (P < 0.05). Furthermore, the relative abundance of Clostridium XIVa (+181%) and colonic butyrate concentration (+53%) were greater in LF + DON compared with DON (P < 0.05). CONCLUSIONS Our study highlights a promising antimycotoxin approach using LF to alleviate DON-induced intestinal dysfunction by modulating the mitogen-activated protein kinase pathway and gut microbial community in mice.
Collapse
Affiliation(s)
- Ping Hu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Qiufang Zong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Yahui Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Haotian Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - YaYa Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Fang Gu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Abdelkareem A Ahmed
- Department of Veterinary Biomedical Sciences, Botswana University of Agriculture and Agriculture and Natural Resources, Ebele, Gaborone, Botswana; Biomedical Research Institute, Darfur University College, Nyala, Sudan
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, PR China.
| |
Collapse
|
19
|
Abdel Baky NA, Al-Najjar AH, Elariny HA, Sallam AS, Mohammed AA. Pramipexole and Lactoferrin ameliorate Cyclophosphamide-Induced haemorrhagic cystitis via targeting Sphk1/S1P/MAPK, TLR-4/NF-κB, and NLRP3/caspase-1/IL-1β signalling pathways and modulating the Nrf2/HO-1 pathway. Int Immunopharmacol 2022; 112:109282. [DOI: 10.1016/j.intimp.2022.109282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/30/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
|
20
|
Ulluwishewa D, Mullaney J, Adam K, Claycomb R, Anderson RC. A bioactive bovine whey protein extract improves intestinal barrier function in vitro. JDS COMMUNICATIONS 2022; 3:387-392. [PMID: 36465501 PMCID: PMC9709612 DOI: 10.3168/jdsc.2022-0245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/30/2022] [Indexed: 06/17/2023]
Abstract
The human intestine plays an important role as a barrier against the ingress of pathogens and other harmful antigens. Accordingly, proper regulation of the intestinal barrier is essential for optimal health. Intestinal barrier function is regulated in part by the interactions between dietary compounds and the intestinal immune system. Bioactive whey proteins from bovine milk (such as lactoferrin, lactoperoxidase, and immunoglobulins) are known to exert a range of physiological functions, including modulation of the immune system, and thus have the potential to regulate intestinal barrier function. While the effects of individual whey proteins on intestinal barrier function have been studied to some extent, less is known about the potentially synergistic properties of whey protein mixtures. Here we investigated the effects of a bioactive bovine whey protein (BWP) extract containing all whey proteins with an isoelectric point >6.8 on intestinal barrier function in vitro. Intestinal epithelial cell (Caco-2) monolayers were treated with BWP before measuring the barrier integrity over 48 h by means of trans-epithelial electrical resistance (TEER). Treatment of epithelial monolayers with 1 mg/mL BWP resulted in an increase in TEER compared with untreated epithelial monolayers. To determine whether BWP could mitigate immune-mediated intestinal barrier dysfunction, we challenged differentiated Caco-2 cell monolayers with tumor necrosis factor α (TNFα) to obtain an in vitro model of a "leaky" intestinal epithelium. The TNFα challenge led to a decrease in TEER over time across untreated control monolayers, indicating a loss of barrier function. This loss of barrier function was mitigated in monolayers treated with 1 mg/mL BWP, but not monolayers treated with the equivalent amount of lactoferrin present in 1 mg/mL BWP. These data suggest that naturally co-occurring bioactive proteins together may enhance intestinal barrier integrity and protect against inflammation-induced barrier dysfunction to a greater extent than lactoferrin alone. Further work is required to determine the key proteins and protein combinations within BWP, and the mechanisms through which BWP modulates intestinal barrier function.
Collapse
Affiliation(s)
- Dulantha Ulluwishewa
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North 4474, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Jane Mullaney
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North 4474, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
- High-Value Nutrition National Science Challenge, Auckland 1142, New Zealand
| | | | | | - Rachel C. Anderson
- AgResearch Ltd., Te Ohu Rangahau Kai, Palmerston North 4474, New Zealand
| |
Collapse
|
21
|
Moysidou CM, Withers AM, Nisbet AJ, Price DRG, Bryant CE, Cantacessi C, Owens RM. Investigation of Host-Microbe-Parasite Interactions in an In Vitro 3D Model of the Vertebrate Gut. Adv Biol (Weinh) 2022; 6:e2200015. [PMID: 35652159 DOI: 10.1002/adbi.202200015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/23/2022] [Indexed: 01/28/2023]
Abstract
In vitro models of the gut-microbiome axis are in high demand. Conventionally, intestinal monolayers grown on Transwell setups are used to test the effects of commensals/pathogens on the barrier integrity, both under homeostatic and pathophysiological conditions. While such models remain valuable for deepening the understanding of host-microbe interactions, often, they lack key biological components that mediate this intricate crosstalk. Here, a 3D in vitro model of the vertebrate intestinal epithelium, interfaced with immune cells surviving in culture for over 3 weeks, is developed and applied to proof-of-concept studies of host-microbe interactions. More specifically, the establishment of stable host-microbe cocultures is described and functional and morphological changes in the intestinal barrier induced by the presence of commensal bacteria are shown. Finally, evidence is provided that the 3D vertebrate gut models can be used as platforms to test host-microbe-parasite interactions. Exposure of gut-immune-bacteria cocultures to helminth "excretory/secretory products" induces in vivo-like up-/down-regulation of certain cytokines. These findings support the robustness of the modular in vitro cell systems for investigating the dynamics of host-microbe crosstalk and pave the way toward new approaches for systems biology studies of pathogens that cannot be maintained in vitro, including parasitic helminths.
Collapse
Affiliation(s)
- Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Aimee M Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| | - Alasdair J Nisbet
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Daniel R G Price
- Moredun Research Institute, Pentlands Science Park, Edinburgh, EH26 0PZ, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, Cambridge Veterinary School, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Philippa Fawcett Drive, West Cambridge Site, CB3 0AS, UK
| |
Collapse
|
22
|
Various Coated Barrier Membranes for Better Guided Bone Regeneration: A Review. COATINGS 2022. [DOI: 10.3390/coatings12081059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A good barrier membrane is one of the important factors for effective guided bone/tissue regeneration (GBR/GTR) in the case of periodontal bone defects. Several methods are being discussed to overcome and improve the shortcomings of commercially available membranes. One of the methods is to coat the membrane with bioactive materials. In this study, 41 studies related to coated membranes for GBR/GTR published in the last 5 years were reviewed. These studies reported coating the membrane with various bioactive materials through different techniques to improve osteogenesis, antimicrobial properties, and physical/mechanical properties. The reported studies have been classified and discussed based on the purpose of coating. The goal of the most actively studied research on coating or surface modification of membranes is to improve new bone formation. For this purpose, calcium phosphate, bioactive glass, polydopamine, osteoinduced drugs, chitosan, platelet-rich fibrin, enamel matrix derivatives, amelotin, hyaluronic acid, tantalum, and copper were used as membrane coating materials. The paradigm of barrier membranes is changing from only inert (or biocompatible) physical barriers to bioactive osteo-immunomodulatory for effective guided bone and tissue regeneration. However, there is a limitation that there exists only a few clinical studies on humans to date. Efforts are needed to implement the use of coated membranes from the laboratory bench to the dental chair unit. Further clinical studies are needed in the patients’ group for long-term follow-up to confirm the effect of various coating materials.
Collapse
|
23
|
Wells JM, Gao Y, de Groot N, Vonk MM, Ulfman L, van Neerven RJJ. Babies, Bugs, and Barriers: Dietary Modulation of Intestinal Barrier Function in Early Life. Annu Rev Nutr 2022; 42:165-200. [PMID: 35697048 DOI: 10.1146/annurev-nutr-122221-103916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The intestinal barrier is essential in early life to prevent infection, inflammation, and food allergies. It consists of microbiota, a mucus layer, an epithelial layer, and the immune system. Microbial metabolites, the mucus, antimicrobial peptides, and secretory immunoglobulin A (sIgA) protect the intestinal mucosa against infection. The complex interplay between these functionalities of the intestinal barrier is crucial in early life by supporting homeostasis, development of the intestinal immune system, and long-term gut health. Exclusive breastfeeding is highly recommended during the first 6 months. When breastfeeding is not possible, milk-based infant formulas are the only safe alternative. Breast milk contains many bioactive components that help to establish the intestinal microbiota and influence the development of the intestinal epithelium and the immune system. Importantly, breastfeeding lowers the risk for intestinal and respiratory tract infections. Here we review all aspects of intestinal barrier function and the nutritional components that impact its functionality in early life, such as micronutrients, bioactive milk proteins, milk lipids, and human milk oligosaccharides. These components are present in breast milk and can be added to milk-based infant formulas to support gut health and immunity. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jerry M Wells
- Host Microbe Interactomics, Wageningen University and Research, Wageningen, The Netherlands
| | - Yifan Gao
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands
| | | | | | | | - R J Joost van Neerven
- Cell Biology and Immunology, Wageningen University and Research, Wageningen, The Netherlands.,FrieslandCampina, Amersfoort, The Netherlands;
| |
Collapse
|
24
|
Fan L, Yao Q, Wu H, Wen F, Wang J, Li H, Zheng N. Protective effects of recombinant lactoferrin with different iron saturations on enteritis injury in young mice. J Dairy Sci 2022; 105:4791-4803. [DOI: 10.3168/jds.2021-21428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 02/16/2022] [Indexed: 01/18/2023]
|
25
|
The Protective Effects of Lactoferrin on Aflatoxin M1-Induced Compromised Intestinal Integrity. Int J Mol Sci 2021; 23:ijms23010289. [PMID: 35008712 PMCID: PMC8745159 DOI: 10.3390/ijms23010289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 11/17/2022] Open
Abstract
Aflatoxin M1 (AFM1), the only toxin with maximum residue levels in milk, has adverse effects on the intestinal barrier, resulting in intestinal inflammatory disease. Lactoferrin (LF), one of the important bioactive proteins in milk, performs multiple biological functions, but knowledge of the protective effects of LF on the compromised intestinal barrier induced by AFM1 has not been investigated. In the present study, results using Balb/C mice and differentiated Caco-2 cells showed that LF intervention decreased AFM1-induced increased intestinal permeability, improved the protein expression of claudin-3, occludin and ZO-1, and repaired the injured intestinal barrier. The transcriptome and proteome were used to clarify the underlying mechanisms. It was found that LF reduced the intestinal barrier dysfunction caused by AFM1 and was associated with intestinal cell survival related pathways, such as cell cycle, apoptosis and MAPK signaling pathway and intestinal integrity related pathways including endocytosis, tight junction, adherens junction and gap junction. The cross-omics analysis suggested that insulin receptor (INSR), cytoplasmic FMR1 interacting protein 2 (CYFIP2), dedicator of cytokinesis 1 (DOCK1) and ribonucleotide reductase regulatory subunit M2 (RRM2) were the potential key regulators as LF repaired the compromised intestinal barrier. These findings indicated that LF may be an alternative treatment for the compromised intestinal barrier induced by AFM1.
Collapse
|
26
|
Lactoferrin Alleviated AFM1-Induced Apoptosis in Intestinal NCM 460 Cells through the Autophagy Pathway. Foods 2021; 11:foods11010023. [PMID: 35010149 PMCID: PMC8750231 DOI: 10.3390/foods11010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Aflatoxin M1 (AFM1) is the only mycotoxin with maximum residue limit in milk, which may result in serious human diseases. On the contrary, lactoferrin (Lf) is an active protein with multiple functions. Studies have confirmed that Lf has a powerful potential to protect the intestines, but the influence of Lf on mycotoxins is not clear. This study aims to explore whether Lf can protect the cytotoxicity induced by AFM1, and determine the underlying mechanisms in human normal colonic epithelial NCM460 cells. The results indicated that AFM1 decreased the cell viability, and increased the levels of apoptosis and autophagy of NCM460 cells. Lf can alleviate the cytotoxicity induced by AFM1 through enhancing cell viability, significantly down-regulated the expression of apoptotic genes and proteins (BAX, caspase3, caspase9, caspase3, and caspase9), and regulated the gene and protein expression of autophagy factors (Atg5, Atg7, Atg12, Beclin1, ULK1, ULK2, LC3, and p62). Furthermore, interference of the key gene Atg5 of autophagy can reduce AFM1-induced apoptosis, which is consistent with the role of Lf, implying that Lf may protect AFM1-induced intestinal injury by inhibiting excessive autophagy-mediated apoptosis. Taken together, our data indicated that Lf has a mitigating effect on apoptosis induced by AFM1 through the autophagy pathway.
Collapse
|
27
|
Rahman S, Ghiboub M, Donkers JM, van de Steeg E, van Tol EAF, Hakvoort TBM, de Jonge WJ. The Progress of Intestinal Epithelial Models from Cell Lines to Gut-On-Chip. Int J Mol Sci 2021; 22:ijms222413472. [PMID: 34948271 PMCID: PMC8709104 DOI: 10.3390/ijms222413472] [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: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
Over the past years, several preclinical in vitro and ex vivo models have been developed that helped to understand some of the critical aspects of intestinal functions in health and disease such as inflammatory bowel disease (IBD). However, the translation to the human in vivo situation remains problematic. The main reason for this is that these approaches fail to fully reflect the multifactorial and complex in vivo environment (e.g., including microbiota, nutrition, and immune response) in the gut system. Although conventional models such as cell lines, Ussing chamber, and the everted sac are still used, increasingly more sophisticated intestinal models have been developed over the past years including organoids, InTESTine™ and microfluidic gut-on-chip. In this review, we gathered the most recent insights on the setup, advantages, limitations, and future perspectives of most frequently used in vitro and ex vivo models to study intestinal physiology and functions in health and disease.
Collapse
Affiliation(s)
- Shafaque Rahman
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
- Department of Pediatric Gastroenterology and Nutrition, Amsterdam University Medical Centers, Emma Children’s Hospital, 1105 AZ Amsterdam, The Netherlands
| | - Joanne M. Donkers
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Evita van de Steeg
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Eric A. F. van Tol
- The Netherlands Organization for Applied Scientific Research (TNO), 3704 HE Zeist, The Netherlands; (J.M.D.); (E.v.d.S.); (E.A.F.v.T.)
| | - Theodorus B. M. Hakvoort
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology, Endocrinology and Metabolism, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, 1105 BK Amsterdam, The Netherlands; (S.R.); (M.G.); (T.B.M.H.)
- Department of Surgery, University of Bonn, 53113 Bonn, Germany
- Correspondence:
| |
Collapse
|
28
|
Lactotransferrin Downregulation Serves as a Potential Predictor for the Therapeutic Effectiveness of mTOR Inhibitors in the Metastatic Clear Cell Renal Cell Carcinoma without PTEN Mutation. Biomedicines 2021; 9:biomedicines9121896. [PMID: 34944711 PMCID: PMC8698394 DOI: 10.3390/biomedicines9121896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/05/2021] [Accepted: 12/11/2021] [Indexed: 11/29/2022] Open
Abstract
Approximately 30% of clear cell renal cell carcinoma (ccRCC) patients develop metastatic spread at the first diagnosis. Therefore, identifying a useful biomarker to predict ccRCC metastasis or therapeutic effectiveness in ccRCC patients is urgently needed. Previously, we demonstrated that lactotransferrin (LTF) downregulation enhanced the metastatic potential of ccRCC. Here, we show that LTF expression conversely associates with the mTORC1 activity as simulated by gene set enrichment analysis (GSEA). Moreover, Western blot analyses revealed that the LTF knockdown promoted, but the inclusion of recombinant human LTF protein suppressed, the phosphorylation of Akt/mTOR proteins in the detected ccRCC cells. Kaplan–Meier analyses demonstrated that the signature of combining an upregulated mTORC1 activity with a downregulated LTF expression referred to a worse overall and progression-free survival probabilities and associated with distant cancer metastasis in TCGA ccRCC patients. Furthermore, we found that the LTF-suppressed Akt/mTOR activation triggered an increased formation of autophagy in the highly metastatic ccRCC cells. The addition of autophagy inhibitor 3-methyadenine restored the LTF-suppressed cellular migration ability of highly metastatic ccRCC cells. Receiver operating characteristic (ROC) analyses showed that the expression of the LTF and MTORC1 gene set, not the autophagy gene set, could be the useful biomarkers to predict 5-year overall survival rate and cancer progression in ccRCC patients. Significantly, the signature of combining mTORC1 upregulation and LTF downregulation was shown as an independent prognostic factor in a multivariate analysis under the progression-free survival condition using the TCGA ccRCC database. Finally, the treatment with mTOR inhibitor rapamycin predominantly reduced the formation of autophagy and ultimately mitigated the cellular migration ability of ccRCC cells with LTF knockdown. Our findings suggest that LTF downregulation is a biomarker for guiding the use of mTOR inhibitors to combat metastatic ccRCC in the clinic.
Collapse
|
29
|
Liu N, Feng G, Zhang X, Hu Q, Sun S, Sun J, Sun Y, Wang R, Zhang Y, Wang P, Li Y. The Functional Role of Lactoferrin in Intestine Mucosal Immune System and Inflammatory Bowel Disease. Front Nutr 2021; 8:759507. [PMID: 34901112 PMCID: PMC8655231 DOI: 10.3389/fnut.2021.759507] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), is one of the main types of intestinal inflammatory diseases with intestine mucosal immune disorder. Intestine mucosal immune system plays a remarkable and important role in the etiology and pathogenesis of IBD. Therefore, understanding the intestine mucosal immune mechanism is a key step to develop therapeutic interventions for IBD. Intestine mucosal immune system and IBD are influenced by various factors, such as inflammation, gut permeability, gut microbiota, and nutrients. Among these factors, emerging evidence show that nutrients play a key role in inflammation activation, integrity of intestinal barrier, and immune cell modulation. Lactoferrin (LF), an iron-binding glycoprotein belonging to transferrin family, is a dietary bioactive component abundantly found in mammalian milk. Notably, LF has been reported to perform diverse biological functions including antibacterial activity, anti-inflammatory activity, intestinal barrier protection, and immune cell modulation, and is involved in maintaining intestine mucosal immune homeostasis. The improved understanding of the properties of LF in intestine mucosal immune system and IBD will facilitate its application in nutrition, clinical medicine, and health. Herein, this review outlines the recent advancements on LF as a potential therapeutic intervention for IBD associated with intestine mucosal immune system dysfunction. We hope this review will provide a reference for future studies and lay a theoretical foundation for LF-based therapeutic interventions for IBD by understanding the particular effects of LF on intestine mucosal immune system.
Collapse
Affiliation(s)
- Ning Liu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Gang Feng
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Xiaoying Zhang
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Qingjuan Hu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Jiaqi Sun
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Yanan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yan Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
30
|
Galla R, Grisenti P, Farghali M, Saccuman L, Ferraboschi P, Uberti F. Ovotransferrin Supplementation Improves the Iron Absorption: An In Vitro Gastro-Intestinal Model. Biomedicines 2021; 9:biomedicines9111543. [PMID: 34829772 PMCID: PMC8615417 DOI: 10.3390/biomedicines9111543] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
Transferrins constitute the most important iron regulation system in vertebrates and some invertebrates. Soluble transferrins, such as bovine lactoferrin and hen egg white ovotransferrin, are glycoproteins with a very similar structure with lobes that complex with iron. In this in vitro study, a comparison of bovine lactoferrin and ovotransferrin was undertaken to confirm the comparability of biological effects. An in vitro gastric barrier model using gastric epithelial cells GTL-16 and an in vitro intestinal barrier model using CaCo-2 cells was employed to evaluate iron absorption and barrier integrity. An analysis of the molecular pathways involving DMT-1 (divalent metal transporter 1), ferritin and ferroportin was also carried out. These in vitro data demonstrate the activity of both 15% saturated and 100% saturated ovotransferrin on the iron regulation system. Compared with the commercial bovine lactoferrin, both 15% saturated and 100% saturated ovotransferrin were found to act in a more physiological manner. Based on these data, it is possible to hypothesise that ovotransferrin may be an excellent candidate for iron supplementation in humans; in particular, 15% saturated ovotransferrin is the overall best performing product. In vivo studies should be performed to confirm this in vitro data.
Collapse
Affiliation(s)
- Rebecca Galla
- Laboratory Physiology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (R.G.); (M.F.); (L.S.)
| | - Paride Grisenti
- Bioseutica B.V., Landbouwweg 83, 3899 BD Zeewolde, The Netherlands;
| | - Mahitab Farghali
- Laboratory Physiology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (R.G.); (M.F.); (L.S.)
| | - Laura Saccuman
- Laboratory Physiology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (R.G.); (M.F.); (L.S.)
| | - Patrizia Ferraboschi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Saldini 50, 20133 Milan, Italy;
| | - Francesca Uberti
- Laboratory Physiology, Department of Translational Medicine, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy; (R.G.); (M.F.); (L.S.)
- Correspondence: ; Tel.: +39-03-2166-0653
| |
Collapse
|
31
|
Influence of iron binding in the structural stability and cellular internalization of bovine lactoferrin. Heliyon 2021; 7:e08087. [PMID: 34632151 PMCID: PMC8487029 DOI: 10.1016/j.heliyon.2021.e08087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/20/2021] [Accepted: 09/26/2021] [Indexed: 02/05/2023] Open
Abstract
Lactoferrin (Lf) is an iron-binding glycoprotein and a component of many external secretions with a wide diversity of functions. Structural studies are important to understand the mechanisms employed by Lf to exert so varied functions. Here, we used guanidine hydrochloride and high hydrostatic pressure to cause perturbations in the structure of bovine Lf (bLf) in apo and holo (unsaturated and iron-saturated, respectively) forms, and analyzed conformational changes by intrinsic and extrinsic fluorescence spectroscopy. Our results showed that the iron binding promotes changes on tertiary structure of bLf and increases its structural stability. In addition, we evaluated the effects of bLf structural change on the kinetics of bLf internalization in Vero cells by confocal fluorescence microscopy, and observed that the holo form was faster than the apo form. This finding may indicate that structural changes promoted by iron binding may play a key role in the intracellular traffic of bLf. Altogether, our data improve the comprehension of bLf stability and uptake, adding knowledge to its potential use as a biopharmaceutical.
Collapse
|
32
|
Zheng J, Wang H, Deng Z, Shan Y, Lü X, Zhao X. Structure and biological activities of glycoproteins and their metabolites in maintaining intestinal health. Crit Rev Food Sci Nutr 2021:1-16. [PMID: 34619993 DOI: 10.1080/10408398.2021.1987857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Glycoproteins formed by covalent links between oligosaccharide and polypeptides are abundant in various food sources. They are less sensitivity to gastrointestinal enzymes, and hence many of them undergo fermentation in the colon by microorganisms. Therefore, the confer various health benefits on the intestinal ecosystem. However, the current understanding of the effect of glycoproteins on intestinal microorganisms and gut health is limited. This is probably due to their heterogeneous structures and complex metabolic programming patterns. The structure and biological activities of glycoproteins and their microbial metabolites were summarized in this review. The metabolic pathways activated by intestinal bacteria were then discussed in relation to their potential benefits on gut health. Food-derived glycoproteins and their metabolites improve gut health by regulating the intestinal bacteria and improving intestinal barrier function, thereby amplifying immune response. The data reviewed here show that food-derived glycoproteins are promising candidates for preventing various gastrointestinal diseases. Further studies should explore the interaction mechanisms between intestinal microorganisms and host metabolites.
Collapse
Affiliation(s)
- Jiaqi Zheng
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Haotian Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Zhanfei Deng
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Yuanyuan Shan
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling, PR China
| | - Xue Zhao
- Department of Nursing, Shandong College of Traditional Chinese Medicine, Yantai, PR China
| |
Collapse
|
33
|
Felix K, Tobias S, Jan H, Nicolas S, Michael M. Measurements of transepithelial electrical resistance (TEER) are affected by junctional length in immature epithelial monolayers. Histochem Cell Biol 2021; 156:609-616. [PMID: 34459960 PMCID: PMC8695537 DOI: 10.1007/s00418-021-02026-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/26/2022]
Abstract
The measurement of transepithelial electrical resistance (TEER) is a common technique to determine the barrier integrity of epithelial cell monolayers. However, it is remarkable that absolute TEER values of similar cell types cultured under comparable conditions show an immense heterogeneity. Based on previous observations, we hypothesized that the heterogeneity of absolute TEER measurements can not only be explained by maturation of junctional proteins but rather by dynamics in the absolute length of cell junctions within monolayers. Therefore, we analyzed TEER in epithelial cell monolayers of Caco2 cells during their differentiation, with special emphasis on both changes in the junctional complex and overall cell morphology within monolayers. We found that in epithelial Caco2 monolayers TEER increased until confluency, then decreased for some time, which was then followed by an additional increase during junctional differentiation. In contrast, permeability of macromolecules measured at different time points as 4 kDA fluorescein isothiocyanate (FITC)-dextran flux across monolayers steadily decreased during this time. Detailed analysis suggested that this observation could be explained by alterations of junctional length along the cell borders within monolayers during differentiation. In conclusion, these observations confirmed that changes in cell numbers and consecutive increase of junctional length have a critical impact on TEER values, especially at stages of early confluency when junctions are immature.
Collapse
Affiliation(s)
- Kannapin Felix
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberduerrbacherstrasse 6, 97080, Würzburg, Germany
| | - Schmitz Tobias
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Roentgenring 11, 97070, Würzburg, Germany
| | - Hansmann Jan
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Roentgenring 11, 97070, Würzburg, Germany.,Translational Center Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Schlegel Nicolas
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberduerrbacherstrasse 6, 97080, Würzburg, Germany
| | - Meir Michael
- Department of General, Visceral, Vascular and Pediatric Surgery, University Hospital Würzburg, Oberduerrbacherstrasse 6, 97080, Würzburg, Germany.
| |
Collapse
|
34
|
Wang W, Cheng Z, Wang X, An Q, Huang K, Dai Y, Meng Q, Zhang Y. Lactoferrin, a Critical Player in Neonate Intestinal Development: RHLF may be a Good Choice in Formula. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8726-8736. [PMID: 34323077 DOI: 10.1021/acs.jafc.1c03129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Lactoferrin (LF) is a bioactive glycoprotein in human milk and has positive effects on neonates. The LF knockout mouse model was generated as a mother mouse that provided LF-free milk. The intestinal development of suckling neonates drinking normal milk and LF-free milk was studied. The results showed that the intestinal density, maturity, and barrier integrity of mice drinking LF-free milk were lower than those of mice drinking normal milk. Therefore, the importance of adding lactoferrin to the human formula is considered. Human lactoferrin (HLF), bovine lactoferrin (BLF), and recombinant HLF (RHLF) were used to compare their functional impact on Caco-2 cell lines. Cell proliferation, differentiation, the establishment of the intestinal barrier, and protective effects on lipopolysaccharide injury were detected. Our results showed that RHLF exhibited more similar functions to HLF than BLF and showed the combined advantages of HLF and BLF in promoting the establishment of the intestinal barrier. This study emphasizes the important role of LF in neonatal intestinal development and provides a theoretical basis for the availability of RHLF.
Collapse
Affiliation(s)
- Wenli Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Zhimei Cheng
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xiong Wang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qin An
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Kunlun Huang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs, Beijing 100083, P. R. China
| | - Yunping Dai
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Qingyong Meng
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Yali Zhang
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
- Key Laboratory of Safety Assessment of Genetically Modified Organism (Food Safety), The Ministry of Agriculture and Rural Affairs, Beijing 100083, P. R. China
| |
Collapse
|
35
|
Goulding DA, Vidal K, Bovetto L, O'Regan J, O'Brien NM, O'Mahony JA. The impact of thermal processing on the simulated infant gastrointestinal digestion, bactericidal and anti-inflammatory activity of bovine lactoferrin - An in vitro study. Food Chem 2021; 362:130142. [PMID: 34087706 DOI: 10.1016/j.foodchem.2021.130142] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/13/2021] [Accepted: 05/16/2021] [Indexed: 02/07/2023]
Abstract
Lactoferrin (LF) is a multifunctional glycoprotein which, when thermally processed, undergoes significant physicochemical changes. The link between such changes and the bioactivity of LF is not well characterised and requires much research. In this work, bovine LF solutions (1%, w/v, protein, pH 7) were thermally processed using high temperature short time conditions (72, 80, 85 or 95 °C with 15 s holding times). Following this, it was shown that LF and heat induced LF aggregates were largely resistant to simulated infant gastric, but not intestinal, digestion. Also, the efficacy of LF bactericidal activity, and inhibition of lipopolysaccharide-induced NF-κB activation were negatively impacted by thermal processing. This study confirmed that the efficacy of LF bio-functionalities was affected by the extent of heat-induced changes in protein structure whereby processing conditions of least severity (i.e. pasteurisation) had the least impact on bioactivity.
Collapse
Affiliation(s)
- David A Goulding
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - Karine Vidal
- Nestlé Research, Nestlé Institute of Health Sciences, Vers-chez-les-Blanc, CH-1000 Lausanne 26, Switzerland
| | - Lionel Bovetto
- Nestlé Research, Nestlé Institute of Material Sciences, Vers-chez-les-Blanc, CH-1000 Lausanne 26, Switzerland
| | - Jonathan O'Regan
- Nestlé Development Centre Nutrition, Askeaton, Co. Limerick, Ireland
| | - Nora M O'Brien
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland
| | - James A O'Mahony
- School of Food and Nutritional Sciences, University College Cork, Cork, Ireland.
| |
Collapse
|
36
|
Lactoferrin as a regenerative agent: The old-new panacea? Pharmacol Res 2021; 167:105564. [PMID: 33744427 DOI: 10.1016/j.phrs.2021.105564] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/01/2021] [Accepted: 03/15/2021] [Indexed: 01/17/2023]
Abstract
Lactoferrin (Lf) possesses various biological properties and therapeutic potentials being a perspective anti-inflammatory, antibacterial, antiviral, antioxidant, antitumor, and immunomodulatory agent. A significant body of literature has also demonstrated that Lf modulates regenerative processes in different anatomical structures, such as bone, cartilage, skin, mucosa, cornea, tendon, vasculature, and adipose tissue. Hence, this review collected and analyzed the data on the regenerative effects of Lf, as well as paid specific attention to their molecular basis. Furthermore, tissue and condition-specific activities of different Lf types as well as problems of their delivery to the targeted organs were discussed. The authors strongly hope that this review will stimulate researchers to focus on the highlighted topics thus accelerating the progress of Lf's wider clinical application.
Collapse
|
37
|
Hering NA, Luettig J, Jebautzke B, Schulzke JD, Rosenthal R. The Punicalagin Metabolites Ellagic Acid and Urolithin A Exert Different Strengthening and Anti-Inflammatory Effects on Tight Junction-Mediated Intestinal Barrier Function In Vitro. Front Pharmacol 2021; 12:610164. [PMID: 33776763 PMCID: PMC7987831 DOI: 10.3389/fphar.2021.610164] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/04/2021] [Indexed: 12/31/2022] Open
Abstract
Scope: Ellagitannins are polyphenols found in numerous fruits, nuts and seeds. The elagitannin punicalagin and its bioactive metabolites ellagic acid and urolithins are discussed to comprise a high potential for therapeutically or preventive medical application such as in intestinal diseases. The present study characterizes effects of punicalagin, ellagic acid and urolithin A on intestinal barrier function in the absence or presence of the proinflammatory cytokine tumor necrosis factor-α (TNFα). Methods and Results: Transepithelial resistance (TER), fluorescein and ion permeability, tight junction protein expression and signalling pathways were examined in Caco-2 and HT-29/B6 intestinal epithelial cell models. Punicalagin had less or no effects on barrier function in both cell models. Ellagic acid was most effective in ileum-like Caco-2 cells, where it increased TER and reduced fluorescein and sodium permeabilities. This was paralleled by myosin light chain kinase two mediated expression down-regulation of claudin-4, -7 and -15. Urolithin A impeded the TNFα-induced barrier loss by inhibition of claudin-1 and -2 protein expression upregulation and claudin-1 delocalization in HT-29/B6. Conclusion: Ellagic acid and urolithin A affect intestinal barrier function in distinct ways. Ellagic acid acts preventive by strengthening the barrier per se, while urolithin A protects against inflammation-induced barrier dysfunction.
Collapse
Affiliation(s)
- Nina A Hering
- Department of General and Visceral Surgery, Charité - Universitätsmedizin Berlin, Berlin, Germasny
| | - Julia Luettig
- Institute of Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Britta Jebautzke
- Institute of Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jörg D Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Rita Rosenthal
- Institute of Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
38
|
Advances in modelling the human microbiome-gut-brain axis in vitro. Biochem Soc Trans 2021; 49:187-201. [PMID: 33544117 PMCID: PMC7924999 DOI: 10.1042/bst20200338] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/23/2020] [Accepted: 01/15/2021] [Indexed: 02/06/2023]
Abstract
The human gut microbiome has emerged as a key player in the bidirectional communication of the gut–brain axis, affecting various aspects of homeostasis and pathophysiology. Until recently, the majority of studies that seek to explore the mechanisms underlying the microbiome–gut–brain axis cross-talk, relied almost exclusively on animal models, and particularly gnotobiotic mice. Despite the great progress made with these models, various limitations, including ethical considerations and interspecies differences that limit the translatability of data to human systems, pushed researchers to seek for alternatives. Over the past decades, the field of in vitro modelling of tissues has experienced tremendous growth, thanks to advances in 3D cell biology, materials, science and bioengineering, pushing further the borders of our ability to more faithfully emulate the in vivo situation. The discovery of stem cells has offered a new source of cells, while their use in generating gastrointestinal and brain organoids, among other tissues, has enabled the development of novel 3D tissues that better mimic the native tissue structure and function, compared with traditional assays. In parallel, organs-on-chips technology and bioengineered tissues have emerged as highly promising alternatives to animal models for a wide range of applications. Here, we discuss how recent advances and trends in this area can be applied in host–microbe and host–pathogen interaction studies. In addition, we highlight paradigm shifts in engineering more robust human microbiome-gut-brain axis models and their potential to expand our understanding of this complex system and hence explore novel, microbiome-based therapeutic approaches.
Collapse
|
39
|
Wang MY, Li Y, Gao M, Song LW, Xu M, Zhao XL, Jia Y, Zhao M, Sun YY, Hu HL. Effects of subacute ruminal acidosis on colon epithelial morphological structure, permeability, and expression of key tight junction proteins in dairy goats. J Dairy Sci 2021; 104:4260-4270. [PMID: 33485680 DOI: 10.3168/jds.2020-18738] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022]
Abstract
The hindgut epithelial barrier plays an important role in maintaining absorption and immune homeostasis in ruminants. However, little information is available on changes in colon epithelial barrier structure and function following grain-induced subacute ruminal acidosis (SARA). The objective of this study was to investigate the effects of grain-induced SARA on colon epithelial morphological structure, permeability, and gene expression involved in epithelial barrier function. Twelve mid-lactating (136 ± 2 d in milk; milk yield = 1.68 ± 0.15 kg/d) Saanen dairy goats with 62.13 ± 4.76 kg of body weight were randomly divided into either the control (CON) treatment (n = 6) or SARA treatment (n = 6). The CON goats were fed a basal diet with a nonfiber carbohydrates to neutral detergent fiber ratio of 1.15 for 60 d. The SARA goats were fed 4 diets with increasing nonfiber carbohydrates to neutral detergent fiber ratio at 1.15, 1.49, 2.12, and 2.66 to induce SARA, with each diet (referred to as period) being fed for 15 d, including 12 d for adaptation and 3 d for sampling. Continuous ruminal pH recordings were used to diagnose the severity of SARA. Additionally, colonic tissues were collected to evaluate the epithelial morphological structure, permeability, and expression of tight junction proteins using transmission electron microscopy, Ussing chamber, quantitative real-time PCR, and Western blotting. Profound disruption in the colonic epithelium was mainly manifested as the electron density of tight junctions decreased, intercellular space widened, and mitochondria swelled in SARA goats. Colon epithelial short-circuit current, tissue conductance, and the mucosal-to-serosal flux of fluorescein isothiocyanate-dextran 4 kDa were increased and potential difference was decreased in SARA goats compared with CON goats. Subacute ruminal acidosis increased mRNA and protein expression levels of CLDN1 and OCLN in the colonic epithelium. Overall, the data of the present study demonstrate that SARA can impair the barrier function of the colonic epithelium at both structural and functional levels, which is associated with severe epithelial structural damage and increased permeability and changes in the expression of tight junction proteins.
Collapse
Affiliation(s)
- M Y Wang
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China; College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, P.R. China; College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Y Li
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China; College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, P.R. China
| | - M Gao
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China
| | - L W Song
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China
| | - M Xu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, P.R. China
| | - X L Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Y Jia
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, P.R. China
| | - M Zhao
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China
| | - Y Y Sun
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, P.R. China
| | - H L Hu
- Institute of Animal Nutrition and Feed, Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010031, P.R. China.
| |
Collapse
|
40
|
Cutone A, Ianiro G, Lepanto MS, Rosa L, Valenti P, Bonaccorsi di Patti MC, Musci G. Lactoferrin in the Prevention and Treatment of Intestinal Inflammatory Pathologies Associated with Colorectal Cancer Development. Cancers (Basel) 2020; 12:E3806. [PMID: 33348646 PMCID: PMC7766217 DOI: 10.3390/cancers12123806] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
The connection between inflammation and cancer is well-established and supported by genetic, pharmacological and epidemiological data. The inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, have been described as important promoters for colorectal cancer development. Risk factors include environmental and food-borne mutagens, dysbalance of intestinal microbiome composition and chronic intestinal inflammation, with loss of intestinal epithelial barrier and enhanced cell proliferation rate. Therapies aimed at shutting down mucosal inflammatory response represent the foundation for IBDs treatment. However, when applied for long periods, they can alter the immune system and promote microbiome dysbiosis and carcinogenesis. Therefore, it is imperative to find new safe substances acting as both potent anti-inflammatory and anti-pathogen agents. Lactoferrin (Lf), an iron-binding glycoprotein essential in innate immunity, is generally recognized as safe and used as food supplement due to its multifunctionality. Lf possesses a wide range of immunomodulatory and anti-inflammatory properties against different aseptic and septic inflammatory pathologies, including IBDs. Moreover, Lf exerts anti-adhesive, anti-invasive and anti-survival activities against several microbial pathogens that colonize intestinal mucosa of IBDs patients. This review focuses on those activities of Lf potentially useful for the prevention/treatment of intestinal inflammatory pathologies associated with colorectal cancer development.
Collapse
Affiliation(s)
- Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | | | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| |
Collapse
|
41
|
Lee J, Lee J, Lee S, Ahmad T, Madhurakkat Perikamana SK, Kim EM, Lee SW, Shin H. Bioactive Membrane Immobilized with Lactoferrin for Modulation of Bone Regeneration and Inflammation. Tissue Eng Part A 2020; 26:1243-1258. [DOI: 10.1089/ten.tea.2020.0015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Jinkyu Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Jinki Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sangmin Lee
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Taufiq Ahmad
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sajeesh Kumar Madhurakkat Perikamana
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Eun Mi Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
| | - Sang Won Lee
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
| | - Heungsoo Shin
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, Hanyang University, Seoul, Republic of Korea
- BK21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, Seoul, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
42
|
Rofeal MG, Elzoghby AO, Helmy MW, Khalil R, Khairy H, Omar S. Dual Therapeutic Targeting of Lung Infection and Carcinoma Using Lactoferrin-Based Green Nanomedicine. ACS Biomater Sci Eng 2020; 6:5685-5699. [DOI: 10.1021/acsbiomaterials.0c01095] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Marian G. Rofeal
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria 21521, Egypt
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Ahmed O. Elzoghby
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Industrial Pharmacy, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Maged W. Helmy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Damanhur University, Damanhur 22511, Egypt
| | - Rowaida Khalil
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria 21521, Egypt
| | - Heba Khairy
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria 21521, Egypt
| | - Sanaa Omar
- Department of Botany and Microbiology, Faculty of Science, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
43
|
Hu P, Zhao F, Wang J, Zhu W. Early-life lactoferrin intervention modulates the colonic microbiota, colonic microbial metabolites and intestinal function in suckling piglets. Appl Microbiol Biotechnol 2020; 104:6185-6197. [DOI: 10.1007/s00253-020-10675-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/22/2020] [Accepted: 05/10/2020] [Indexed: 12/13/2022]
|
44
|
Panahipour L, Biasi MD, Bokor TS, Thajer A, Haiden N, Gruber R. Milk lactoperoxidase decreases ID1 and ID3 expression in human oral squamous cell carcinoma cell lines. Sci Rep 2020; 10:5836. [PMID: 32246075 PMCID: PMC7125221 DOI: 10.1038/s41598-020-62390-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 03/12/2020] [Indexed: 01/07/2023] Open
Abstract
Milk consumption may modify the risk of squamous cell carcinoma. The role of milk to modulate the gene expression in oral squamous cell carcinoma cells has not been investigated so far. Here, HSC2 oral squamous carcinoma cells were exposed to an aqueous fraction of human milk and a whole-genome array was performed. Among the genes that were significantly reduced by human and cow milk were the DNA-binding protein inhibitor 1 (ID1), ID3 and Distal-Less Homeobox 2 (DLX2) in HSC2 cells. Also, in TR146 oral squamous carcinoma cells, there was a tendency towards a decreased gene expression. Upon size fractionation, lactoperoxidase but not lactoferrin and osteopontin was identified to reduce ID1 and ID3 in HSC2 cells. Dairy products and hypoallergenic infant formula failed to decrease the respective genes. These data suggest that milk can reduce the expression of transcription factors in oral squamous carcinoma cells.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Maria De Biasi
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Theresa Sophia Bokor
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria
| | - Alexandra Thajer
- Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, Sensengasse 2a, 1090, Vienna, Austria. .,Department of Periodontology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010, Bern, Switzerland. .,Austrian Cluster for Tissue Regeneration, Donaueschingenstraße 13, 1200, Vienna, Austria.
| |
Collapse
|
45
|
Hirahatake KM, Bruno RS, Bolling BW, Blesso C, Alexander LM, Adams SH. Dairy Foods and Dairy Fats: New Perspectives on Pathways Implicated in Cardiometabolic Health. Adv Nutr 2020; 11:266-279. [PMID: 31555799 PMCID: PMC7442361 DOI: 10.1093/advances/nmz105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/13/2022] Open
Abstract
Low-fat and nonfat dairy products have been promoted as part of a healthy dietary pattern by both US dietary guidelines and professional organizations for several decades. The basis for this recommendation stems in part from the putative negative cardiometabolic effects associated with saturated fat consumption. However, as nutrition research has shifted from a single nutrient to a whole-food/dietary pattern approach, the role of dairy foods and dairy fat in the diet-disease relationship is being reexamined. Most observational and experimental evidence does not support a detrimental relationship between full-fat dairy intake and cardiometabolic health, including risks of cardiovascular disease and type 2 diabetes. Indeed, an expanded understanding of the dairy food matrix and the bioactive properties of dairy fats and other constituents suggests a neutral or potentially beneficial role in cardiometabolic health. To consider how consuming dairy foods, including full-fat dairy, is associated with cardiometabolic health, this review provides an innovative perspective on mechanisms that link dairy consumption to 3 main biological systems at the core of metabolic health, the gastrointestinal, hepatic, and vascular systems.
Collapse
Affiliation(s)
- Kristin M Hirahatake
- Department of Epidemiology, College of Health Sciences, University of California, Irvine, CA, USA
| | - Richard S Bruno
- Human Nutrition Program, Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, OH, USA
| | - Bradley W Bolling
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Christopher Blesso
- Department of Nutritional Sciences, College of Agriculture, Health and Natural Resources, University of Connecticut, Storrs, CT, USA
| | - Lacy M Alexander
- Department of Kinesiology, College of Health and Human Development, The Pennsylvania State University, State College, PA, USA
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, AR, USA,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Address correspondence to SHA (e-mail: )
| |
Collapse
|
46
|
Hu P, Zhao F, Wang J, Zhu W. Lactoferrin attenuates lipopolysaccharide-stimulated inflammatory responses and barrier impairment through the modulation of NF-κB/MAPK/Nrf2 pathways in IPEC-J2 cells. Food Funct 2020; 11:8516-8526. [DOI: 10.1039/d0fo01570a] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lactoferrin attenuated LPS-induced inflammatory responsesviainhibiting NF-κB/MAPK pathways in IPEC-J2 cells.
Collapse
Affiliation(s)
- Ping Hu
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Fangzhou Zhao
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| |
Collapse
|
47
|
Hering NA, Fromm A, Bücker R, Gorkiewicz G, Zechner E, Högenauer C, Fromm M, Schulzke JD, Troeger H. Tilivalline- and Tilimycin-Independent Effects of Klebsiella oxytoca on Tight Junction-Mediated Intestinal Barrier Impairment. Int J Mol Sci 2019; 20:E5595. [PMID: 31717457 PMCID: PMC6888351 DOI: 10.3390/ijms20225595] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Klebsiella oxytoca causes antibiotic-associated hemorrhagic colitis and diarrhea. This was attributed largely to its secreted cytotoxins tilivalline and tilimycin, inductors of epithelial apoptosis. To study whether Klebsiella oxytoca exerts further barrier effects, T84 monolayers were challenged with bacterial supernatants derived from tilivalline/tilimycin-producing AHC6 or its isogeneic tilivalline/tilimycin-deficient strain Mut-89. Both preparations decreased transepithelial resistance, enhanced fluorescein and FITC-dextran-4kDa permeabilities, and reduced expression of barrier-forming tight junction proteins claudin-5 and -8. Laser scanning microscopy indicated redistribution of both claudins off the tight junction region in T84 monolayers as well as in colon crypts of mice infected with AHC6 or Mut-89, indicating that these effects are tilivalline/tilimycin-independent. Furthermore, claudin-1 was affected, but only in a tilivalline/tilimycin-dependent manner. In conclusion, Klebsiella oxytoca induced intestinal barrier impairment by two mechanisms: the tilivalline/tilimycin-dependent one, acting by increasing cellular apoptosis and a tilivalline/tilimycin-independent one, acting by weakening the paracellular pathway through the tight junction proteins claudin-5 and -8.
Collapse
Affiliation(s)
- Nina A. Hering
- Medical Department of General, Visceral and Vascular Surgery, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Anja Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany; (A.F.); (R.B.); (M.F.); (J.-D.S.)
| | - Roland Bücker
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany; (A.F.); (R.B.); (M.F.); (J.-D.S.)
| | - Gregor Gorkiewicz
- Institute of Pathology, Medical University of Graz, A-8036 Graz, Austria;
| | - Ellen Zechner
- BioTechMed-Graz, Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria;
| | - Christoph Högenauer
- Division of Gastroenterology and Hepatology, Medical University of Graz, A-8036 Graz, Austria;
| | - Michael Fromm
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany; (A.F.); (R.B.); (M.F.); (J.-D.S.)
| | - Jörg-Dieter Schulzke
- Institute of Clinical Physiology/Nutritional Medicine, Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany; (A.F.); (R.B.); (M.F.); (J.-D.S.)
| | - Hanno Troeger
- Medical Department, Division of Gastroenterology, Infectiology and Rheumatology, Charité – Universitätsmedizin Berlin, 12203 Berlin, Germany;
| |
Collapse
|
48
|
Pang X, Tong Y, Li F, Wei X, Chen X, Liu J, Chen D. Expression and characterization of human lactoferrin with tandem zinc finger protein in Chlamydomonas reinhardtii. ALGAL RES 2019. [DOI: 10.1016/j.algal.2019.101635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Hu P, Zhao F, Zhu W, Wang J. Effects of early-life lactoferrin intervention on growth performance, small intestinal function and gut microbiota in suckling piglets. Food Funct 2019; 10:5361-5373. [PMID: 31393487 DOI: 10.1039/c9fo00676a] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The early postnatal stage is a critical period for suckling animals in developing intestinal function and stabilizing gut microbiota. Lactoferrin (LF) plays a critical role in promoting gut development and regulating gut microbiota. This study investigates the impact of early-life lactoferrin (LF) intervention on the growth performance, small intestinal function and gut microbiota in suckling piglets. Sixty suckling piglets (1.51 ± 0.05 kg) obtained from six sows (10 piglets per litter) were assigned to a control (CON) group and an LF group in each litter, which were sow-fed. Piglets in the LF group were orally administered 8-12 mL LF solution (0.5 g per kg body weight per day) for a week, and piglets in the CON group were orally administered the same dose of physiological saline. Six piglets (n = 6) from each group were euthanized on days 8 and 21. The early-life LF intervention increased growth performance, with higher villi height of the jejunum and greater disaccharidase activity of the jejunum and ileum (P < 0.05). Diarrhoea incidence decreased in the LF group from day 1 to day 7 (P < 0.05). Urinary lactulose-mannitol ratios decreased in the LF group, whereas the gene and protein expressions of jejunal occludin increased in the LF group on day 8 and day 21, and higher gene and protein levels of ileal occludin were observed on day 8 (P < 0.05). Additionally, the LF piglets had lower concentrations of IL-1β and TNF-α, and higher concentration of IL-10 in the jejunum (P < 0.05). For the ileum, higher concentration of IL-10 and lower concentration of TNF-α were observed in the LF group (P < 0.05). LF piglets had a greater abundance of Lactobacillus and lower abundance of Veillonella and Escherichia-Shigella in the jejunum on day 8 (P < 0.05). In the ileum, the abundance of Actinobacillus was decreased in the LF piglets on day 8 and day 21 (P < 0.05). The early-life LF intervention enhanced the growth performance and decreased diarrhoea incidence in the suckling piglets by promoting the development of intestinal function and changing the microbiota in the small intestine.
Collapse
Affiliation(s)
- Ping Hu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Fangzhou Zhao
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Laboratory of Gastrointestinal Microbiology, National Experimental Teaching Demonstration Center of Animal Science, College of Animal Science and Technology, Nanjing Agricultural University, Jiangsu Province 210095, P. R. China. and Laboratory of Gastrointestinal Microbiology, Xiaolingwei Street, Weigang 1, Nanjing 210095, China
| |
Collapse
|
50
|
Skrypnik K, Bogdański P, Sobieska M, Suliburska J. The effect of multistrain probiotic supplementation in two doses on iron metabolism in obese postmenopausal women: a randomized trial. Food Funct 2019; 10:5228-5238. [PMID: 31384878 DOI: 10.1039/c9fo01006h] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Obesity leads to detrimental abnormalities of iron (Fe) metabolism. So far, studies have shown that single-strain probiotic supplementation ameliorates the gut microbiota quality disrupted in the obese and improves Fe homeostasis. The effect of multistrain probiotic supplementation and its dose-dependence in obese postmenopausal women remain unknown. The study aimed to investigate the effect of multistrain probiotic supplementation in two doses on selected parameters of Fe metabolism in obese postmenopausal female patients. Three groups of obese postmenopausal women, 30 subjects each, received nine-strain oral probiotic supplement at a daily dose of 2.5 × 109 CFU (LD group), 1 × 1010 CFU (HD group), or placebo for 12 weeks (ClinicalTrails.gov no: NCT03100162). After the intervention, the hair Fe content was lower in both supplemented groups compared to the baseline, the serum zinc (Zn) concentration was higher in the LD group and lower in the HD group vs. the baseline, and the serum erythroferrone (FAM) concentration was lower in the HD group and serum ferritin (FE) concentration was higher in the LD group vs. the baseline. In the whole study population after the completion of the intervention hair Zn correlated positively with serum HEPC and FAM and negatively with serum FE. Hair Fe correlated negatively with serum FE. It is concluded that multistrain probiotic supplementation may influence iron metabolism in obese postmenopausal female patients.
Collapse
Affiliation(s)
- Katarzyna Skrypnik
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego St. 31, Poznan, 60-624, Poland.
| | | | | | | |
Collapse
|