51
|
Rahman MM, Takashima S, Kamatari YO, Shimizu K, Okada A, Inoshima Y. Comprehensive Proteomic Analysis Revealed a Large Number of Newly Identified Proteins in the Small Extracellular Vesicles of Milk from Late-Stage Lactating Cows. Animals (Basel) 2021; 11:2506. [PMID: 34573471 PMCID: PMC8470060 DOI: 10.3390/ani11092506] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/16/2021] [Accepted: 08/23/2021] [Indexed: 01/23/2023] Open
Abstract
Bovine milk contains small extracellular vesicles (sEVs) that provide proteins, miRNAs, mRNAs, DNAs, and lipids to target cells and play a role in intracellular communications. Previous studies have characterized proteins in milk sEVs from early- and mid-stage lactation. However, the proteins in milk sEVs from late-stage lactation are mostly unexplored. The aim of this study was to determine the proteomic profile of milk sEVs from late-stage lactating cows. A comprehensive nanoliquid chromatography-tandem mass spectrometry (nanoLC-MS/MS) approach was carried out to reveal the proteins in milk sEVs. Additionally, bioinformatics analysis was carried out to interpret the molecular signatures of newly identified proteins in milk sEVs from three late-stage lactating cows. NanoLC-MS/MS analysis revealed a total of 2225 proteins in milk sEVs from cows. Notably, after comparing these identified proteins with previously deposited datasets of proteins in bovine milk sEVs, 429 proteins were detected as newly identified. Bioinformatic analysis indicated that these newly identified proteins in milk sEVs were engaged in a diverse range of molecular phenomena relevant to mammary gland physiology, milk production, immunity, and immune response. These findings suggest that the newly identified proteins could expand the inventory application of molecular cargos, nutritional status, and immune modulation of sEVs in milk during the late-stage lactation.
Collapse
Affiliation(s)
- Md. Matiur Rahman
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.S.); (A.O.)
- Department of Medicine, Faculty of Veterinary, Animal and Biomedical Sciences, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Shigeo Takashima
- Life Science Research Center, Division of Genomics Research, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
| | - Yuji O. Kamatari
- Life Science Research Center, Division of Instrumental Analysis, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
| | - Kaori Shimizu
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.S.); (A.O.)
| | - Ayaka Okada
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.S.); (A.O.)
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), 1-1 Yanagido, Gifu 501-1193, Japan
| | - Yasuo Inoshima
- The United Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
- Laboratory of Food and Environmental Hygiene, Cooperative Department of Veterinary Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan; (K.S.); (A.O.)
- Education and Research Center for Food Animal Health, Gifu University (GeFAH), 1-1 Yanagido, Gifu 501-1193, Japan
- Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
52
|
Desai N, Gadeval A, Kathar U, Sengupta P, Kalia K, Tekade RK. Emerging roles and biopharmaceutical applications of milk derived exosomes. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
53
|
Ong SL, Blenkiron C, Haines S, Acevedo-Fani A, Leite JAS, Zempleni J, Anderson RC, McCann MJ. Ruminant Milk-Derived Extracellular Vesicles: A Nutritional and Therapeutic Opportunity? Nutrients 2021; 13:2505. [PMID: 34444665 PMCID: PMC8398904 DOI: 10.3390/nu13082505] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Milk has been shown to contain a specific fraction of extracellular particles that are reported to resist digestion and are purposefully packaged with lipids, proteins, and nucleic acids to exert specific biological effects. These findings suggest that these particles may have a role in the quality of infant nutrition, particularly in the early phase of life when many of the foundations of an infant's potential for health and overall wellness are established. However, much of the current research focuses on human or cow milk only, and there is a knowledge gap in how milk from other species, which may be more commonly consumed in different regions, could also have these reported biological effects. Our review provides a summary of the studies into the extracellular particle fraction of milk from a wider range of ruminants and pseudo-ruminants, focusing on how this fraction is isolated and characterised, the stability and uptake of the fraction, and the reported biological effects of these fractions in a range of model systems. As the individual composition of milk from different species is known to differ, we propose that the extracellular particle fraction of milk from non-traditional and minority species may also have important and distinct biological properties that warrant further study.
Collapse
Affiliation(s)
- Siew Ling Ong
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1051, New Zealand;
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1051, New Zealand
| | - Stephen Haines
- Beyond Food Innovation Centre of Excellence, AgResearch Ltd., Lincoln 7674, New Zealand;
| | - Alejandra Acevedo-Fani
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Juliana A. S. Leite
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - Rachel C. Anderson
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Mark J. McCann
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| |
Collapse
|
54
|
Jia W, Zhang R, Zhu Z, Shi L. LC-Q-Orbitrap HRMS-based proteomics reveals potential nutritional function of goat whey fraction. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
55
|
Jiang X, You L, Zhang Z, Cui X, Zhong H, Sun X, Ji C, Chi X. Biological Properties of Milk-Derived Extracellular Vesicles and Their Physiological Functions in Infant. Front Cell Dev Biol 2021; 9:693534. [PMID: 34249944 PMCID: PMC8267587 DOI: 10.3389/fcell.2021.693534] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are released by all cells under pathological and physiological conditions. EVs harbor various biomolecules, including protein, lipid, non-coding RNA, messenger RNA, and DNA. In 2007, mRNA and microRNA (miRNA) carried by EVs were found to have regulatory functions in recipient cells. The biological function of EVs has since then increasingly drawn interest. Breast milk, as the most important nutritional source for infants, contains EVs in large quantities. An increasing number of studies have provided the basis for the hypothesis associated with information transmission between mothers and infants via breast milk-derived EVs. Most studies on milk-derived EVs currently focus on miRNAs. Milk-derived EVs contain diverse miRNAs, which remain stable both in vivo and in vitro; as such, they can be absorbed across different species. Further studies have confirmed that miRNAs derived from milk-derived EVs can resist the acidic environment and enzymatic hydrolysis of the digestive tract; moreover, they can be absorbed by intestinal cells in infants to perform physiological functions. miRNAs derived from milk EVs have been reported in the maturation of immune cells, regulation of immune response, formation of neuronal synapses, and development of metabolic diseases such as obesity and diabetes. This article reviews current status and advances in milk-derived EVs, including their history, biogenesis, molecular contents, and biological functions. The effects of milk-derived EVs on growth and development in both infants and adults were emphasized. Finally, the potential application and future challenges of milk-derived EVs were discussed, providing comprehensive understanding and new insight into milk-derived EVs.
Collapse
Affiliation(s)
- Xue Jiang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.,The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Lianghui You
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Zhenxing Zhang
- The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Xianwei Cui
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hong Zhong
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xingzhen Sun
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Chenbo Ji
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xia Chi
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| |
Collapse
|
56
|
Review of Methodological Approaches to Human Milk Small Extracellular Vesicle Proteomics. Biomolecules 2021; 11:biom11060833. [PMID: 34204944 PMCID: PMC8228857 DOI: 10.3390/biom11060833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/27/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022] Open
Abstract
Proteomics can map extracellular vesicles (EVs), including exosomes, across disease states between organisms and cell types. Due to the diverse origin and cargo of EVs, tailoring methodological and analytical techniques can support the reproducibility of results. Proteomics scans are sensitive to in-sample contaminants, which can be retained during EV isolation procedures. Contaminants can also arise from the biological origin of exosomes, such as the lipid-rich environment in human milk. Human milk (HM) EVs and exosomes are emerging as a research interest in health and disease, though the experimental characterization and functional assays remain varied. Past studies of HM EV proteomes have used data-dependent acquisition methods for protein detection, however, improvements in data independent acquisition could allow for previously undetected EV proteins to be identified by mass spectrometry. Depending on the research question, only a specific population of proteins can be compared and measured using isotope and other labelling techniques. In this review, we summarize published HM EV proteomics protocols and suggest a methodological workflow with the end-goal of effective and reproducible analysis of human milk EV proteomes.
Collapse
|
57
|
Sedykh SE, Purvinish LV, Burkova EE, Dmitrenok PS, Vlassov VV, Ryabchikova EI, Nevinsky GA. Analysis of peptides and small proteins in preparations of horse milk exosomes, purified on anti-CD81-Sepharose. Int Dairy J 2021. [DOI: 10.1016/j.idairyj.2021.104994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
58
|
Zeng B, Wang H, Luo J, Xie M, Zhao Z, Chen X, Wang D, Sun J, Xi Q, Chen T, Zhang Y. Porcine Milk-Derived Small Extracellular Vesicles Promote Intestinal Immunoglobulin Production through pIgR. Animals (Basel) 2021; 11:ani11061522. [PMID: 34073819 PMCID: PMC8225040 DOI: 10.3390/ani11061522] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/16/2021] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary As the irreplaceable nutrient source for young mammals, milk has a number of biological functions. Milk derived extracellular vesicles are a recently discovered component of milk and have potential regulatory effects on intestinal health and immunity. In this study, in vivo and in vitro experiments were performed to examine the effects of porcine milk small extracellular vesicles (PM-sEVs) on intestinal immunity. As a result, PM-sEVs promoted intestinal secretory immunoglobulin A (SIgA) levels, and increased the expression levels of the polymeric immunoglobulin receptor (pIgR) both in mice and piglet. We identified circ-XPO4 in PM-sEVs as a crucial circRNA, which promotes the expression of pIgR via the suppression of miR-221-5p in the intestinal cell. In conclusion, our research provides a new understanding of the nutritional physiology of porcine milk in intestinal immunity. Abstract Secretory immunoglobulin A (SIgA) plays an important role in gut acquired immunity and mucosal homeostasis. Breast milk is the irreplaceable nutritional source for mammals after birth. Current studies have shown the potential functional role of milk-derived small extracellular vesicles (sEVs) and their RNAs cargo in intestinal health and immune regulation. However, there is a lack of studies to demonstrate how milk-derived sEVs affect intestinal immunity in recipient. In this study, through in vivo experiments, we found that porcine milk small extracellular vesicles (PM-sEVs) promoted intestinal SIgA levels, and increased the expression levels of polymeric immunoglobulin receptor (pIgR) both in mice and piglet. We examined the mechanism of how PM-sEVs increased the expression level of pIgR in vitro by using a porcine small intestine epithelial cell line (IPEC-J2). Through bioinformatics analysis, dual-luciferase reporter assays, and overexpression or knockdown of the corresponding non-coding RNAs, we identified circ-XPO4 in PM-sEVs as a crucial circRNA, which leads to the expression of pIgR via the suppression of miR-221-5p in intestinal cells. Importantly, we also observed that oral administration of PM-sEVs increased the level of circ-XPO4 and decreased the level of miR-221-5p in small intestine of piglets, indicating that circRNAs in milk-derived sEVs act as sponge for miRNAs in recipients. This study, for the first time, reveals that PM-sEVs have a capacity to stimulate intestinal SIgA production by delivering circRNAs to receptors and sponging the recipient’s original miRNAs, and also provides valuable data for insight into the role and mechanism of animal milk sEVs in intestinal immunity.
Collapse
|
59
|
Gao HN, Hu H, Wen PC, Lian S, Xie XL, Song HL, Yang ZN, Ren FZ. Yak milk-derived exosomes alleviate lipopolysaccharide-induced intestinal inflammation by inhibiting PI3K/AKT/C3 pathway activation. J Dairy Sci 2021; 104:8411-8424. [PMID: 34001362 DOI: 10.3168/jds.2021-20175] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/05/2021] [Indexed: 01/05/2023]
Abstract
Intestinal epithelial cells (IEC) are important parts of the mucosal barrier, whose function can be impaired upon various injury factors such as lipopolysaccharide. Although food-derived exosomes are preventable against intestinal barrier injuries, there have been few studies on the effect of yak milk-derived exosomes and the underlying mechanism that remains poorly understood. This study aimed to characterize the effect of exosomal proteins derived from yak and cow milk on the barrier function of IEC-6 treated with lipopolysaccharide and the relevant mechanism involved. Proteomics study revealed 392 differentially expressed proteins, with 58 higher expressed and 334 lower expressed in yak milk-derived exosomes than those in cow exosomes. Additionally, the top 20 proteins with a relatively consistent higher expression in yak milk exosomes than cow milk exosomes were identified. Protein CD46 was found to be a regulator for alleviating inflammatory injury of IEC-6. In vitro assay of the role of yak milk exosomes on survival of IEC-6 in inflammation by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide assay confirmed the effectiveness of yak milk exosomes to increase IEC-6 survival up to 18% for 12 h compared with cow milk exosomes (up to 12%), indicating a therapeutic effect of yak milk exosomes in the prevention of intestinal inflammation. Furthermore, yak and cow milk exosomes were shown to activate the PI3K/AKT/C3 signaling pathway, thus promoting IEC-6 survival. Our findings demonstrated an important relationship between yak and cow milk exosomes and intestinal inflammation, facilitating further understanding of the mechanisms of inflammation-driven epithelial homeostasis. Interestingly, compared with cow milk exosomes, yak milk exosomes activated the PI3K/AKT/C3 signaling pathway more to lower the incidence and severity of intestine inflammation, which might represent a potential innovative therapeutic option for intestinal inflammation.
Collapse
Affiliation(s)
- H N Gao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - H Hu
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, China
| | - P C Wen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - S Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - X L Xie
- Treasure of Tibet Yak Dairy Co., Ltd., Lhasa 610000, China
| | - H L Song
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China
| | - Z N Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - F Z Ren
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
60
|
Extracellular vesicles isolated from milk can improve gut barrier dysfunction induced by malnutrition. Sci Rep 2021; 11:7635. [PMID: 33828139 PMCID: PMC8026962 DOI: 10.1038/s41598-021-86920-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 03/18/2021] [Indexed: 02/01/2023] Open
Abstract
Malnutrition impacts approximately 50 million children worldwide and is linked to 45% of global mortality in children below the age of five. Severe acute malnutrition (SAM) is associated with intestinal barrier breakdown and epithelial atrophy. Extracellular vesicles including exosomes (EVs; 30-150 nm) can travel to distant target cells through biofluids including milk. Since milk-derived EVs are known to induce intestinal stem cell proliferation, this study aimed to examine their potential efficacy in improving malnutrition-induced atrophy of intestinal mucosa and barrier dysfunction. Mice were fed either a control (18%) or a low protein (1%) diet for 14 days to induce malnutrition. From day 10 to 14, they received either bovine milk EVs or control gavage and were sacrificed on day 15, 4 h after a Fluorescein Isothiocyanate (FITC) dose. Tissue and blood were collected for histological and epithelial barrier function analyses. Mice fed low protein diet developed intestinal villus atrophy and barrier dysfunction. Despite continued low protein diet feeding, milk EV treatment improved intestinal permeability, intestinal architecture and cellular proliferation. Our results suggest that EVs enriched from milk should be further explored as a valuable adjuvant therapy to standard clinical management of malnourished children with high risk of morbidity and mortality.
Collapse
|
61
|
Go G, Jeon J, Lee G, Lee JH, Lee SH. Bovine milk extracellular vesicles induce the proliferation and differentiation of osteoblasts and promote osteogenesis in rats. J Food Biochem 2021; 45:e13705. [PMID: 33748986 DOI: 10.1111/jfbc.13705] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/19/2021] [Accepted: 03/05/2021] [Indexed: 12/20/2022]
Abstract
Bone is constantly balanced between the formation of new bone by osteoblasts and the absorption of old bone by osteoclasts. To promote bone growth and improve bone health, it is necessary to promote the proliferation and differentiation of osteoblasts. Although bovine milk is known to exert a beneficial effect on bone formation, the study on the effect of bovine milk extracellular vesicles (EVs) on osteogenesis in osteoblasts is limited. In this study, we demonstrated that bovine milk EVs promoted the proliferation of human osteogenic Saos-2 cells by increasing the expression of cell cycle-related proteins. In addition, bovine milk EVs also induced the differentiation of Saos-2 cells by increasing the expression of RUNX2 and Osterix which are key transcription factors for osteoblast differentiation. Oral administration of milk EVs did not cause toxicity in Sprague-Dawley rats. Furthermore, milk EVs promoted longitudinal bone growth and increased the bone mineral density of the tibia. Our findings suggest that milk EVs could be a safe and powerful applicant for enhancing osteogenesis. PRACTICAL APPLICATIONS: Until now, calcium and vitamin D have been prescribed to promote bone formation or to prevent bone diseases such as osteoporosis. Recently, several studies to find bioactive molecules that regulate cellular functions of osteoblasts or osteoclasts are actively underway. Milk basic proteins and lactoferrin present in milk are known to promote bone formation, but they exist in small quantities and the isolation of these proteins is complicated making mass production difficult. Recently, it has been found that milk contains large quantities of EVs, and that they promote bone formation. Studies on the effect of Milk EVs on osteoblasts during osteogenesis will help in the development of biomaterials for osteogenesis.
Collapse
Affiliation(s)
- Gyeongyun Go
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea.,Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | | | - Gaeun Lee
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea.,Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, Republic of Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Republic of Korea.,Department of Oral Anatomy, College of Dentistry, Dankook University, Cheonan, Republic of Korea.,Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Sang Hun Lee
- Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea.,Department of Biochemistry, BK21FOUR Project2, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Stembio, Ltd, Asan, Republic of Korea.,Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| |
Collapse
|
62
|
Jan AT, Rahman S, Badierah R, Lee EJ, Mattar EH, Redwan EM, Choi I. Expedition into Exosome Biology: A Perspective of Progress from Discovery to Therapeutic Development. Cancers (Basel) 2021; 13:1157. [PMID: 33800282 PMCID: PMC7962655 DOI: 10.3390/cancers13051157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are membrane-enclosed distinct cellular entities of endocytic origin that shuttle proteins and RNA molecules intercellularly for communication purposes. Their surface is embossed by a huge variety of proteins, some of which are used as diagnostic markers. Exosomes are being explored for potential drug delivery, although their therapeutic utilities are impeded by gaps in knowledge regarding their formation and function under physiological condition and by lack of methods capable of shedding light on intraluminal vesicle release at the target site. Nonetheless, exosomes offer a promising means of developing systems that enable the specific delivery of therapeutics in diseases like cancer. This review summarizes information on donor cell types, cargoes, cargo loading, routes of administration, and the engineering of exosomal surfaces for specific peptides that increase target specificity and as such, therapeutic delivery.
Collapse
Affiliation(s)
- Arif Tasleem Jan
- School of Biosciences and Biotechnology, Baba Ghulam Shah Badshah University, Rajouri 185234, India;
| | - Safikur Rahman
- Department of Botany, MS College, BR Ambedkar Bihar University, Muzaffarpur, Bihar 842001, India;
| | - Raied Badierah
- Biological Sciences Department, Faculty of Science, and Laboratory University Hospital, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (R.B.); (E.H.M.)
| | - Eun Ju Lee
- Department of Medical Biotechnology and Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| | - Ehab H. Mattar
- Biological Sciences Department, Faculty of Science, and Laboratory University Hospital, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (R.B.); (E.H.M.)
| | - Elrashdy M. Redwan
- Biological Sciences Department, Faculty of Science, and Laboratory University Hospital, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia; (R.B.); (E.H.M.)
| | - Inho Choi
- Department of Medical Biotechnology and Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Korea;
| |
Collapse
|
63
|
Han X, Chen L, Hu Z, Chen L, Sun P, Wang Y, Liu Y. Identification of proteins related with pemetrexed resistance by iTRAQ and PRM-based comparative proteomic analysis and exploration of IGF2BP2 and FOLR1 functions in non-small cell lung cancer cells. J Proteomics 2021; 237:104122. [PMID: 33561557 DOI: 10.1016/j.jprot.2021.104122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/17/2020] [Accepted: 01/19/2021] [Indexed: 12/25/2022]
Abstract
Pemetrexed (PEM), a multi-target folate antagonist, has been extensively used for the treatment of non-small cell lung cancer (NSCLC). However, the therapeutic efficacy of PEM is limited by tumor resistance. In this project, iTRAQ and parallel reaction monitoring (PRM)-based LC-MS/MS comparative proteomic analysis was performed to identify protein determinants of PEM resistance in A549/PEM cells versus A549 parental cells. A total of 567 differentially expressed proteins (DEPs) were identified by iTRAQ analysis. The function and classification of DEPs were analyzed through GO and KEGG Pathway databases. Moreover, PRM analysis further validated the expression changes of 14 DEPs identified by iTRAQ analysis. Moreover, insulin-like growth factor (IGF) 2 mRNA-binding protein 2 (IGF2BP2) or folate receptor alpha (FOLR1) knockdown weakened PEM resistance, reduced cell viability and promoted cell apoptosis in A549/PEM cells. IGF2BP2 depletion inhibited cell migration, invasion and epithelial-mesenchymal transition (EMT), while FOLR1 loss had no much effect on cell migration, invasion and EMT in A549/PEM cells. Our study can provide a deep insight into molecular mechanisms of PEM resistance in NSCLC and contribute to the development of more effective therapeutic schedules. SIGNIFICANCE: Our study can provide deeper insight into molecular mechanisms of PEM resistance in NSCLC and contribute to the development of more effective therapeutic schedules.
Collapse
Affiliation(s)
- Xiaobing Han
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China.
| | - Liangfeng Chen
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China
| | - Zhongzhou Hu
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China
| | - Liangxin Chen
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China
| | - Peng Sun
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China
| | - Yujun Wang
- Department of Gastroenterology, Xinyang Central Hospital, Xinyang, 464000, China
| | - Yangyang Liu
- Department of Oncology, Xinyang Central Hospital, Xinyang 464000, China
| |
Collapse
|
64
|
Proteomic profiling of milk small extracellular vesicles from bovine leukemia virus-infected cattle. Sci Rep 2021; 11:2951. [PMID: 33536533 PMCID: PMC7858626 DOI: 10.1038/s41598-021-82598-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Milk small extracellular vesicles (sEV) contain proteins that provide potential information of host physiology and immunology. Bovine leukemia virus (BLV) is an oncogenic virus that causes progressive B-cell lymphosarcoma in cattle. In this study, we aimed to explore the proteomic profile of milk sEV from BLV-infected cattle compared with those from uninfected cattle. Milk sEV were isolated from three BLV-infected and three uninfected cattle. Proteomic analysis was performed by using a comprehensive nanoLC-MS/MS method. Furthermore, gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were used to evaluate the candidates for uniquely or differentially expressed proteins in milk sEV from BLV-infected cattle. Proteomic analysis revealed a total of 1330 common proteins in milk sEV among BLV-infected cattle, whereas 118 proteins were uniquely expressed compared with those from uninfected cattle. Twenty-six proteins in milk sEV were differentially expressed proteins more than two-fold significant difference (p < 0.05) in BLV-infected cattle. GO and KEGG analyses indicated that the candidates for uniquely or differentially expressed proteins in milk sEV had been involved in diverse biological activities including metabolic processes, cellular processes, respond to stimulus, binding, catalytic activities, cancer pathways, focal adhesion, and so on. Taken together, the present findings provided a novel insight into the proteomes of milk sEV from BLV-infected cattle.
Collapse
|
65
|
Civra A, Francese R, Donalisio M, Tonetto P, Coscia A, Sottemano S, Balestrini R, Faccio A, Cavallarin L, Moro GE, Bertino E, Lembo D. Human Colostrum and Derived Extracellular Vesicles Prevent Infection by Human Rotavirus and Respiratory Syncytial Virus in Vitro. J Hum Lact 2021; 37:122-134. [PMID: 33534629 DOI: 10.1177/0890334420988239] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND It is known that breastfeeding protects the infant from enteric and respiratory infections; however, the antiviral properties of human milk against enteric and respiratory viruses are largely unexplored. RESEARCH AIMS To explore the antiviral activity of human preterm colostrum against rotavirus and respiratory syncytial virus and to assess whether the derived extracellular vesicle contribute to this activity. METHODS We used a cross-sectional, prospective two-group non-experimental design. Colostra were collected from mothers of preterm newborns (N = 10) and extracellular vesicles were purified and characterized. The antiviral activity of colostra and derived extracellular vesicles were tested in vitro against rotavirus and respiratory syncytial virus and the step of viral replication inhibited by extracellular vesicles was investigated. RESULTS Each sample of colostrum and colostrum-derived extracellular vesicles had significant antiviral activity with a wide interpersonal variability. Mechanism of action studies demonstrated that extracellular vesicles acted by interfering with the early steps of the viral replicative cycle. CONCLUSION We demonstrated the intrinsic antiviral activity of human colostrum against rotavirus and respiratory syncytial virus and we showed that extracellular vesicles substantially contribute to the overall protective effect. Our results contribute to unravelling novel mechanisms underlying the functional role of human milk as a protective and therapeutic agent in preterm infants.
Collapse
Affiliation(s)
- Andrea Civra
- 93149314 Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Rachele Francese
- 93149314 Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Manuela Donalisio
- 93149314 Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Paola Tonetto
- Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| | - Alessandra Coscia
- Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| | - Stefano Sottemano
- Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| | - Raffaella Balestrini
- National Research Council - Institute for Sustainable Plant Protection (CNR-IPSP), Turin Unit, Italy
| | - Antonella Faccio
- National Research Council - Institute for Sustainable Plant Protection (CNR-IPSP), Turin Unit, Italy
| | - Laura Cavallarin
- 9327 Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, Grugliasco (TO), Italy
| | - Guido E Moro
- Italian Association of Human Milk Banks, Milan, Italy
| | - Enrico Bertino
- Neonatal Care Unit of the University, City of Health and Science Hospital, Turin, Italy
| | - David Lembo
- 93149314 Laboratory of Molecular Virology and Antiviral Research, Department of Clinical and Biological Sciences, University of Turin, Italy
| |
Collapse
|
66
|
Ahn G, Kim YH, Ahn JY. Multifaceted effects of milk-exosomes (Mi-Exo) as a modulator of scar-free wound healing. NANOSCALE ADVANCES 2021; 3:528-537. [PMID: 36131751 PMCID: PMC9419160 DOI: 10.1039/d0na00665c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/26/2020] [Indexed: 05/09/2023]
Abstract
Scar-free treatment is complex involving many cells in the human body but a very elaborate reaction. This process demands regulation of various growth factors on behalf of TGFβ3 around the damaged tissue, and it is also important to protect cells from inflammatory reactions and oxidative stress to avoid abnormalities. Here, we focused on bovine derived milk exosomes (Mi-Exo) and their scar-free healing potential. The physiological properties (size and shape), biological markers (TSG101 and Bta-miR2478) and stability on storage of Mi-Exo were analyzed. Mi-Exo exhibited significant NP (number of Mi-Exo particles)-dependent scavenging activity in ABTS assay. In addition, Mi-Exo suppressed the expression of pro-inflammatory mediators, IL-6 and TNFα, and pro-inflammatory chemokines, COX-2 and iNOS. This study showed that cell migration was significantly inhibited in a Mi-Exo NP-dependent manner. We also evaluated the expression of TGFβ1 and TGFβ3 on the basis of mRNA and protein levels. Furthermore, the role of functional behavior of Mi-Exo in TGFβ1 maturation was explored. This is the first study to demonstrate that Mi-Exo may target the TGFβ signaling pathway, which plays important roles in scar-free wound healing.
Collapse
Affiliation(s)
- Gna Ahn
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| | - Ji-Young Ahn
- School of Biological Sciences, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea
| |
Collapse
|
67
|
WITHDRAWN: Enzymatic hydrolysis effects on immunoreactivity and nutritional quality of natural cow milk. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.110927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
68
|
iTRAQ®-based quantitative proteomics reveals the proteomic profiling of methicillin-resistant Staphylococcus aureus-derived extracellular vesicles after exposure to imipenem. Folia Microbiol (Praha) 2020; 66:221-230. [PMID: 33165807 DOI: 10.1007/s12223-020-00836-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022]
Abstract
This study sought to reveal the proteomic profiling of methicillin-resistant Staphylococcus aureus (MRSA)-derived extracellular vesicles (EVs) after exposure to imipenem. The advanced isobaric tags for relative and absolute quantitation (iTRAQ®) proteomic approach were used to analyze the alterations in MRSA-derived EV protein patterns upon exposure to imipenem. A total of 1260 EV proteins were identified and quantified. Among these, 861 differentially expressed exosome proteins (P < 0.05) were found. Multivariate analysis, Gene Ontology (GO) annotation, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were used to analyze the identified proteins. Enrichment analysis of GO annotations indicated that imipenem primarily regulated the metabolic processes in MRSA. The metabolism of differentially expressed proteins was found to be the most significant in the combined analysis of the KEGG pathway analysis. Based on the results from the STRING analysis, 50S ribosomal protein L16 (RplP) and 30S ribosomal protein S8 (RpsH) were involved in the imipenem-induced MRSA-derived EVs. These results provide vital information on MRSA-derived EVs, increasing our knowledge of the proteome level changes in EVs upon exposure to imipenem. Moreover, these results pave the way for developing novel MRSA treatments.
Collapse
|
69
|
Villatoro AJ, Martín-Astorga MDC, Alcoholado C, Becerra J. Canine colostrum exosomes: characterization and influence on the canine mesenchymal stem cell secretory profile and fibroblast anti-oxidative capacity. BMC Vet Res 2020; 16:417. [PMID: 33138803 PMCID: PMC7607682 DOI: 10.1186/s12917-020-02623-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Background Canine colostrum milk (CCM) is a specific secretion of the mammary gland that is fundamental for the survival of the newborn. CCM has many described components (immunoglobulins, proteins or fat), but its small vesicles, named exosomes, are largely unknown. Results A characterization of CCM exosomes was performed. Exosomes were abundant in CCM and appeared with the characteristic cup-shaped morphology and well-defined round vesicles. The size distribution of exosomes was between 37 and 140 nm, and western blot analysis showed positive expression of specific exosomal markers. Proteomic analysis revealed a total of 826 proteins in exosome cargo. We also found that exosomes modified the proliferation and secretory profiles in canine mesenchymal stem cells derived from bone marrow (cBM-MSCs) and adipose tissue (cAd-MSCs). Additionally, CCM exosomes demonstrated a potent antioxidant effect on canine fibroblasts in culture. Conclusions Our findings highlight, for the first time, the abundant presence of exosomes in CCM and their ability to interact with mesenchymal stem cells (MSCs). The addition of exosomes to two types of MSCs in culture resulted in specific secretory profiles with functions related to angiogenesis, migration and chemotaxis of immune cells. In particular, the cAd-MSCs secretory profile showed higher potential in adipose tissue development and neurogenesis, while cBM-MSC production was associated with immunity, cell mobilization and haematopoiesis. Finally, exosomes also presented antioxidant capacity on fibroblasts against reactive oxygen species activity within the cell, demonstrating their fundamental role in the development and maturation of dogs in the early stages of their life. Supplementary information Supplementary information accompanies this paper at 10.1186/s12917-020-02623-w.
Collapse
Affiliation(s)
- Antonio J Villatoro
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, 29071, Málaga, Spain.,Instituto de Immunología Clínica y Terapia Celular (IMMUNESTEM), Miraflores del Palo, 14, 29018, Málaga, Spain
| | - María Del Carmen Martín-Astorga
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, 29071, Málaga, Spain
| | - Cristina Alcoholado
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, 29071, Málaga, Spain.,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain
| | - José Becerra
- Laboratory of Bioengineering and Tissue Regeneration (LABRET), Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, University of Málaga, IBIMA, 29071, Málaga, Spain. .,Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029, Madrid, Spain. .,Andalusian Centre for Nanomedicine and Biotechnology-BIONAND, Severo Ochoa 35, 29590, Málaga, Spain.
| |
Collapse
|
70
|
Sedykh S, Kuleshova A, Nevinsky G. Milk Exosomes: Perspective Agents for Anticancer Drug Delivery. Int J Mol Sci 2020; 21:E6646. [PMID: 32932782 PMCID: PMC7555228 DOI: 10.3390/ijms21186646] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/05/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Exosomes are biological nanovesicles that participate in intercellular communication by transferring biologically active chemical compounds (proteins, microRNA, mRNA, DNA, and others). Due to their small size (diameter 40-100 nm) and high biological compatibility, exosomes are promising delivery tools in personalized therapy. Because artificial exosome synthesis methods are not developed yet, the urgent task is to develop an effective and safe way to obtain exosomes from natural sources. Milk is the only exosome-containing biological fluid that is commercially available. In this regard, milk exosomes are unique and promising candidates for new therapeutic approaches to treating various diseases, including cancer. The appearance of side effects during the use of cytotoxic and cytostatic agents is among the main problems in cancer chemotherapy. According to this, the targeted delivery of chemotherapeutic agents can be a potential solution to the toxic effect of chemotherapy. The ability of milk exosomes to carry out biologically active substances to the cell makes them promising tools for oral delivery of chemotherapeutic agents. This review is devoted to the methods of milk exosome isolation, their biological components, and prospects for their use in cancer treatment.
Collapse
Affiliation(s)
- Sergey Sedykh
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia; (A.K.); (G.N.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Anna Kuleshova
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia; (A.K.); (G.N.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia; (A.K.); (G.N.)
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
71
|
Shifts in the Holstein dairy cow milk fat globule membrane proteome that occur during the first week of lactation are affected by parity. J Anim Sci Biotechnol 2020; 11:81. [PMID: 32695335 PMCID: PMC7367219 DOI: 10.1186/s40104-020-00478-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022] Open
Abstract
Background The milk fat globule membrane (MFGM) proteomes of colostrum and transition milk are rich sources of proteins that are likely important for neonatal calf health. In addition, characterization of these proteomes could also yield valuable information regarding mammary gland physiology of the early postpartum lactating cow. The objectives of this research were to characterize the MFGM proteomes of colostrum and transition milk through sample collections at four timepoints postpartum, including the first milking (M1, colostrum), second milking (M2, transition milk), fourth milking (M4, transition milk), and fourteenth milking (M14, mature milk), and compare these proteomes between multiparous (MP; n = 10) and primiparous (PP; n = 10) Holstein dairy cows. Isolated MFGM proteins were labeled using Tandem Mass tagging and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Protein identification was completed using MASCOT and Sequest in Proteome Discoverer 2.2. The scaled abundance values were analyzed using PROC MIXED in SAS to determine the effects of milking (MIL), parity (PAR), and MIL × PAR. The adaptive false-discovery rate (FDR)-adjusted P values were determined using PROC MULTTEST. Protein characterization and bioinformatic analysis were completed using a combination of PANTHER, Blast, and Uniprot. Results A total of 104 common proteins were identified in each of the MFGM samples. Statistical analysis revealed that 70.2% of identified proteins were affected by MIL. Of these, 78.1% were lower in M14 compared with M1, including immune-related proteins lactotransferrin, lactadherin and hemopexin. Parity affected 44.2% of proteins. Of the proteins affected by PAR, 84.8% were higher in MP cows compared with PP cows, including apolipoprotein E and histones 2A, 2B, 3, and 4 b. Butyrophilin subfamily 1 member 1A and annexin 5 were higher in samples from PP cows. Milking × parity affected 32.7% of identified proteins, including lactotransferrin, gelsolin, vitamin D binding protein, and S100 proteins. Conclusions This research supports previous findings that the Holstein MFGM proteome changes rapidly during the first week of lactation. In addition, this research identifies the impact of parity on the colostrum and transition milk MFGM proteomes, which may be important for milk-fed calf health or for the identification of protein biomarkers for mammary functionality.
Collapse
|
72
|
Ahmadi M, Rezaie J. Tumor cells derived-exosomes as angiogenenic agents: possible therapeutic implications. J Transl Med 2020; 18:249. [PMID: 32571337 PMCID: PMC7310379 DOI: 10.1186/s12967-020-02426-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis is a multistep process and various molecules are involved in regulating it. Extracellular vesicles are cell-derived particles, secreted from several types of cells and are known to mediate cell-to-cell communication. These vesicles contain different bio-molecules including nucleic acids, proteins, and lipids, which are transported between cells and regulate physiological and pathological conditions in the recipient cell. Exosomes, 30–150 nm extracellular vesicles, and their key roles in tumorigenesis via promoting angiogenesis are of great recent interest. In solid tumors, the suitable blood supply is the hallmark of their progression, growth, and metastasis, so it can be supported by angiogenesis. Tumor cells abundantly release exosomes containing different kinds of biomolecules such as angiogenic molecules that contribute to inducing angiogenesis. These exosomes can be trafficked between tumor cells or between tumor cells and endothelial cells. The protein and nucleic acid cargo of tumor derived-exosomes can deliver to endothelial cells mostly by endocytosis, and then induce angiogenesis. Tumor derived-exosomes can be used as biomarker for cancer diagnosis. Targeting exosome-induced angiogenesis may serve as a promising tool for cancer therapy. Taken together, tumor derived-exosomes are the major contributors in tumor angiogenesis and a supposed target for antiangiogenic therapies. However, further scrutiny is essential to investigate the function of exosomes in tumor angiogenesis and clinical relevance of targeting exosomes for suppressing angiogenesis.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd, 1138, Urmia, 57147, Iran.
| |
Collapse
|
73
|
Munir J, Lee M, Ryu S. Exosomes in Food: Health Benefits and Clinical Relevance in Diseases. Adv Nutr 2020; 11:687-696. [PMID: 31796948 PMCID: PMC7231590 DOI: 10.1093/advances/nmz123] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/14/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022] Open
Abstract
Exosomes are membrane-bound organelles generally secreted by eukaryotic cells that contain mRNAs, microRNAs, and/or proteins. However, recent studies have reported the isolation of these particles from foods such as lemon, ginger, and milk. Owing to their absorption by intestinal cells and further travel via the bloodstream, exosomes can reach distant organs and affect overall health in both infants and adults. The potential role of food-derived exosomes (FDEs) in alleviating diseases, as well as in modulating the gut microbiota has been shown, but the underlying mechanism is still unknown. Moreover, exosomes may provide biocompatible vehicles for the delivery of anti-cancer drugs, such as doxorubicin. Thus, exosomes may allow medical nutritionists and clinicians to develop safe and targeted therapies for the treatment of various pathologies. The present review introduces FDEs and their contents, highlights their role in disease and infant/adult health, and explores their potential use as therapeutic agents.
Collapse
Affiliation(s)
| | - Mihye Lee
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan, Chungcheongnam-do, South Korea
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-bioscience (SIMS), Soonchunhyang University, Cheonan, Chungcheongnam-do, South Korea
| |
Collapse
|
74
|
Yang M, Deng W, Cao X, Wang L, Yu N, Zheng Y, Wu J, Wu R, Yue X. Quantitative Phosphoproteomics of Milk Fat Globule Membrane in Human Colostrum and Mature Milk: New Insights into Changes in Protein Phosphorylation during Lactation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:4546-4556. [PMID: 32208690 DOI: 10.1021/acs.jafc.9b06850] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Phosphorylation is a widespread posttranslational protein modification and is important in various biological processes. However, milk fat globule membrane (MFGM) phosphoproteins have not been explored systematically in human milk. Here, we used quantitative phosphoproteomics to analyze phosphorylation sites in human MFGM proteins and their differences at different stages of lactation; 305 phosphorylation sites on 170 proteins and 269 phosphorylation sites on 170 proteins were identified in colostrum and mature MFGM, respectively. Among these, 71 phosphorylation sites on 48 proteins were differentially expressed between the different stages of lactation. Osteopontin in human MFGM was the most heavily phosphorylated protein, with a total of 39 identified phosphorylation sites. Our results shed light on phosphorylation sites, composition, and biological functions of MFGM phosphoproteins in human colostrum and mature milk, and provide novel insights into the crucial roles of protein phosphorylation during infant development.
Collapse
Affiliation(s)
- Mei Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Wei Deng
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Xueyan Cao
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Lijie Wang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Na Yu
- Liaoning General Fair Testing Company, Ltd, Shenyang 110026, China
| | - Yan Zheng
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, PR China
| |
Collapse
|
75
|
Ye F, Wang Y, He Q, Cui C, Yu H, Lu Y, Zhu S, Xu H, Zhao X, Yin H, Li D, Li H, Zhu Q. Exosomes Transmit Viral Genetic Information and Immune Signals may cause Immunosuppression and Immune Tolerance in ALV-J Infected HD11 cells. Int J Biol Sci 2020; 16:904-920. [PMID: 32140061 PMCID: PMC7053331 DOI: 10.7150/ijbs.35839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/06/2019] [Indexed: 01/13/2023] Open
Abstract
Avian leukosis virus (ALV) is oncogenic retrovirus that not only causes immunosuppression but also enhances the host's susceptibility to secondary infection. Exosomes play vital role in the signal transduction cascades that occur in response to viral infection. We want to explore the function of exosomes in the spread of ALV and the body's subsequent immunological response. RNA-sequencing and the isobaric tags for relative and absolute quantitation (iTRAQ) method were used to detect differentially expressed genes (DEGs) and differentially expressed proteins (DEPs) in exosomes secreted by macrophage cells in response to injection with ALV subgroup J (ALV-J). RNA-sequencing identified 513 DEGs in infected cells, with specific differential regulation in mRNA involved in tight junction signaling, TNF signaling, salmonella infection response, and immune response, among other important cellular processes. Differential regulation was observed in 843 lncRNAs, with particular enrichment in those lncRNA targets involved in Rap1 signaling, HTLV-I infection, tight junction signaling, and other signaling pathways. A total of 50 DEPs were identified in the infected cells by iTRAQ. The proteins enriched are involved in immune response, antigen processing, the formation of both MHC protein and myosin complexes, and transport. Combined analysis of the transcriptome and proteome revealed that there were 337 correlations between RNA and protein enrichment, five of which were significant. Pathways that were enriched on both the RNA and protein levels were involved in pathways in cancer, PI3K-Akt signaling pathway, Endocytosis, Epstein-Barr virus infection. These data show that exosomes are transmitters of intercellular signaling in response to viral infection. Exosomes can carry both viral nucleic acids and proteins, making it possible for exosomes to be involved in the viral infection of other cells and the transmission of immune signals between cells. Our sequencing results confirme previous studies on exosomes and further find exosomes may cause immunosuppression and immune tolerance.
Collapse
Affiliation(s)
- Fei Ye
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China.,Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan, 528231, Guangdong, China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Qijian He
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Can Cui
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Heling Yu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Yuxiang Lu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Shiliang Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Hengyong Xu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Xiaoling Zhao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Huadong Yin
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Diyan Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| | - Hua Li
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China.,Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Foshan University, Foshan, 528231, Guangdong, China
| | - Qing Zhu
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Sichuan, Chengdu, China
| |
Collapse
|
76
|
Metabolomics methods to analyze full spectrum of amino acids in different domains of bovine colostrum and mature milk. Eur Food Res Technol 2019. [DOI: 10.1007/s00217-019-03385-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
77
|
Ye F, He Q, Wang Y, Cui C, Yang F, Luo B, Yin H, Zhao X, Li D, Xu H, Li H, Zhu Q. Data-independent acquisition of the proteomics of spleens from chickens infected by avian leukosis virus. 3 Biotech 2019; 9:332. [PMID: 31475084 DOI: 10.1007/s13205-019-1863-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 08/05/2019] [Indexed: 12/24/2022] Open
Abstract
Immunosuppression caused by avian leukemia virus J subgroup (ALV-J) infection includes atrophy or regeneration disorders of the lymphoid organs, decreased immune response, and termination of B lymphocyte maturation process and inhibition of T-lymphocyte development. The regulatory mechanism of the related resistance genes and protein expression is not clear. While searching for a molecular marker for the immune response to ALV-J infection, we detected differentially expressed proteins (DEPs) of spleens from chicken infected by ALV-J at 15th day and 30th day by the data-independent acquisition technique. Approximately 220 DEPs from the spleens of chickens infected by ALV-J were detected. To find a relatively stable biomarker molecule, we summarized the DEPs at two timepoints and selected activating signal cointegrator 1 complex subunit 3 (ASCC3), TBC1 domain family member 2 (TBC1D2), MHC class II beta chain 1 (BLB2), ensconsin (MAP7), complement component 1 Q subcomponent B chain (C1QB), and Follistatin-like 1 (FSTL1) from both comparisons for protein interaction network analysis. ASCC3, BLB2, C1QB, and FSTL1 were potential biomarkers for the complex infection mechanism of ALV-J and the dynamic immune mechanism of the body.
Collapse
|
78
|
Subedi P, Schneider M, Philipp J, Azimzadeh O, Metzger F, Moertl S, Atkinson MJ, Tapio S. Comparison of methods to isolate proteins from extracellular vesicles for mass spectrometry-based proteomic analyses. Anal Biochem 2019; 584:113390. [PMID: 31401005 DOI: 10.1016/j.ab.2019.113390] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-bound organelles that have generated interest as they reflect the physiological condition of their source. Mass spectrometric (MS) analyses of protein cargo of EVs may lead to the discovery of biomarkers for diseases. However, for a comprehensive MS-based proteomics analysis, an optimal lysis of the EVs is required. Six methods for the protein extraction from EVs secreted by the head and neck cell line BHY were compared. Commercial radioimmunoprecipitation assay (RIPA) buffer outperformed the other buffers investigated in this study (Tris-SDS, Tris-Triton, GuHCl, urea-thiourea, and commercial Cell-lysis buffer). Following lysis with RIPA buffer, 310 proteins and 1469 peptides were identified using LTQ OrbitrapXL mass spectrometer. Among these, 86% of proteins and 72% of peptides were identified in all three replicates. In the case of other buffers, Tris-Triton identified on average 277 proteins, Cell-lysis buffer 257 proteins, and Tris-SDS, GuHCl and urea-thiourea each 267 proteins. In total, 399 proteins including 74 of the top EV markers (Exocarta) were identified, the most of the latter (73) using RIPA. The proteins exclusively identified using RIPA represented all Gene Ontology cell compartments. This study suggests that RIPA is an optimal lysis buffer for EVs in combination with MS.
Collapse
Affiliation(s)
- Prabal Subedi
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany.
| | - Michael Schneider
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Jos Philipp
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Omid Azimzadeh
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Fabian Metzger
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Research Unit Protein Science, Munich, Germany
| | - Simone Moertl
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Michael J Atkinson
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| | - Soile Tapio
- Helmholtz Zentrum München, German Research Centre for Environmental Health GmbH, Institute of Radiation Biology, Neuherberg, Germany
| |
Collapse
|
79
|
Milk proteome from in silico data aggregation allows the identification of putative biomarkers of negative energy balance in dairy cows. Sci Rep 2019; 9:9718. [PMID: 31273261 PMCID: PMC6609625 DOI: 10.1038/s41598-019-46142-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/19/2019] [Indexed: 01/13/2023] Open
Abstract
A better knowledge of the bovine milk proteome and its main drivers is a prerequisite for the modulation of bioactive proteins in milk for human nutrition, as well as for the discovery of biomarkers that are useful in husbandry and veterinary medicine. Milk composition is affected by lactation stage and reflects, in part, the energy balance of dairy cows. We aggregated the cow milk proteins reported in 20 recent proteomics publications to produce an atlas of 4654 unique proteins. A multistep assessment was applied to the milk proteome datasets according to lactation stages and milk fractions, including annotations, pathway analysis and literature mining. Fifty-nine proteins were exclusively detected in milk from early lactation. Among them, we propose six milk proteins as putative biomarkers of negative energy balance based on their implication in metabolic adaptative pathways. These proteins are PCK2, which is a gluconeogenic enzyme; ACAT1 and IVD, which are involved in ketone metabolism; SDHA and UQCRC1, which are related to mitochondrial oxidative metabolism; and LRRC59, which is linked to mammary gland cell proliferation. The cellular origin of these proteins warrants more in-depth research but may constitute part of a molecular signature for metabolic adaptations typical of early lactation.
Collapse
|
80
|
Betker JL, Angle BM, Graner MW, Anchordoquy TJ. The Potential of Exosomes From Cow Milk for Oral Delivery. J Pharm Sci 2019; 108:1496-1505. [PMID: 30468828 PMCID: PMC6788294 DOI: 10.1016/j.xphs.2018.11.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/19/2018] [Accepted: 11/16/2018] [Indexed: 01/23/2023]
Abstract
Many pharmaceuticals must be administered intravenously due to their poor oral bioavailability. In addition to issues associated with sterility and inconvenience, the cost of repeated infusion over a 6-week course of therapy costs the health care system tens of billions of dollars per year. Attempts to improve oral bioavailability have traditionally focused on enhancing drug solubility and membrane permeability, and the use of synthetic nanoparticles has also been investigated. As an alternative strategy, some recent reports have clearly demonstrated that exosomes from cow milk are absorbed from the gastrointestinal tract in humans and could potentially be used for oral delivery of drugs that are traditionally administered intravenously. Our previous work has shown that antibodies are present in exosome preparations, and the current work with milk exosomes suggests that absorption from the gastrointestinal tract occurs via the "neonatal" Fc receptor, FcRn. Furthermore, our results demonstrate that milk exosomes are absorbed from the gut as intact particles that can be modified with ligands to promote retention in target tissues.
Collapse
Affiliation(s)
- Jamie L Betker
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Brittany M Angle
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Michael W Graner
- Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Thomas J Anchordoquy
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045.
| |
Collapse
|
81
|
Jimenez L, Yu H, McKenzie AJ, Franklin JL, Patton JG, Liu Q, Weaver AM. Quantitative Proteomic Analysis of Small and Large Extracellular Vesicles (EVs) Reveals Enrichment of Adhesion Proteins in Small EVs. J Proteome Res 2019; 18:947-959. [PMID: 30608700 DOI: 10.1021/acs.jproteome.8b00647] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) are important mediators of cell-cell communication due to their cargo content of proteins, lipids, and RNAs. We previously reported that small EVs (SEVs) called exosomes promote directed and random cell motility, invasion, and serum-independent growth. In contrast, larger EVs (LEVs) were not active in those assays, but might have unique functional properties. In order to identify protein cargos that may contribute to different functions of SEVs and LEVs, we used isobaric tags for relative and absolute quantitation (iTRAQ)-liquid chromatography (LC) tandem mass spectrometry (MS) on EVs isolated from a colon cancer cell line. Bioinformatics analyses revealed that SEVs are enriched in proteins associated with cell-cell junctions, cell-matrix adhesion, exosome biogenesis machinery, and various signaling pathways. In contrast, LEVs are enriched in proteins associated with ribosome and RNA biogenesis, processing, and metabolism. Western blot analysis of EVs purified from two different cancer cell types confirmed the enrichment of cell-matrix and cell-cell adhesion proteins in SEVs. Consistent with those data, we found that cells exhibit enhanced adhesion to surfaces coated with SEVs compared to an equal protein concentration of LEVs. These data suggest that a major function of SEVs is to promote cellular adhesion.
Collapse
Affiliation(s)
- Lizandra Jimenez
- Department of Cell and Developmental Biology , Vanderbilt University School of Medicine , Nashville , Tennessee 37232 , United States
| | - Hui Yu
- Department of Internal Medicine , University of New Mexico , Albuquerque , New Mexico 87131 , United States
| | - Andrew J McKenzie
- Sarah Cannon Research Institute , Nashville , Tennessee 37203 , United States
| | - Jeffrey L Franklin
- Department of Cell and Developmental Biology , Vanderbilt University School of Medicine , Nashville , Tennessee 37232 , United States.,Department of Medicine , Vanderbilt University Medical Center , Nashville , Tennessee 37212 , United States
| | - James G Patton
- Department of Biological Sciences , Vanderbilt University School of Medicine , Nashville , Tennessee 37212 , United States
| | - Qi Liu
- Department of Biostatistics , Vanderbilt University Medical Center , Nashville , Tennessee 37232 , United States
| | - Alissa M Weaver
- Department of Cell and Developmental Biology , Vanderbilt University School of Medicine , Nashville , Tennessee 37232 , United States.,Department of Pathology, Microbiology and Immunology , Vanderbilt University Medical Center , Nashville , Tennessee 37212 , United States
| |
Collapse
|
82
|
Gao HN, Guo HY, Zhang H, Xie XL, Wen PC, Ren FZ. Yak-milk-derived exosomes promote proliferation of intestinal epithelial cells in an hypoxic environment. J Dairy Sci 2018; 102:985-996. [PMID: 30580945 DOI: 10.3168/jds.2018-14946] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/02/2018] [Indexed: 12/18/2022]
Abstract
Intestinal epithelial cells (IEC) are an important part of the intestinal barrier. Barrier function was disrupted under hypoxia, but milk-derived exosomes can regulate the intestinal barrier function. However, the mechanisms underlying the association between yak milk exosomes and hypoxia in IEC remain poorly understood. In this follow-up study, we proposed an effective optimization method for purifying yak-milk-derived exosomes. The Western blot analyses indicated that the expression of the proteins of the endosomal sorting complexes required for transport (TSG101), proteins of the tetraspanin family (CD63), and heat shock protein 70 (Hsp-70) proteins from yak-milk-derived exosomes were significantly higher than those in cow-milk-derived exosomes. Flow cytometry analysis showed that yak milk had 3.7 times the number of exosomes compared with cow milk. Moreover, we explored whether yak milk exosomes could facilitate intestinal cell survival under hypoxic conditions in vitro. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide results showed that yak-milk-derived exosomes significantly increased survival of IEC-6 cells with rates of up to 29% for cells incubated in hypoxic conditions for 12 h, compared with those of cow-milk-derived exosomes posttreatment (rates of up to 22% for cells incubated in hypoxic conditions for 12 h). Confocal microscopy revealed that the IEC-6 cells uptake more yak-milk-derived exosomes than cow milk in hypoxic conditions. Furthermore, the Western blot analyses indicated that yak-milk-derived exosomes significantly promote oxygen-sensitive prolyl hydroxylase (PHD)-1 expression and decrease the expression of hypoxia-inducible factor-α and its downstream target vascular endothelial growth factor (VEGF) in the IEC-6 cells. Further, yak-milk-derived exosomes significantly inhibited p53 levels. In conclusion, our findings demonstrate that yak-milk-derived exosomes more effectively activate the hypoxia-inducible factor signaling pathway, thus promoting IEC-6 cell survival, which may result in higher hypoxia tolerance than cow-milk-derived exosomes.
Collapse
Affiliation(s)
- H N Gao
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China; Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - H Y Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100094, China
| | - H Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100094, China
| | - X L Xie
- Treasure of Tibet Yak Dairy Co., Ltd., Lhasa, 610000, China
| | - P C Wen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China.
| | - F Z Ren
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China; Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100094, China.
| |
Collapse
|
83
|
Liang X, Han H, Zhao X, Cao X, Yang M, Tao D, Wu R, Yue X. Quantitative analysis of amino acids in human and bovine colostrum milk samples through iTRAQ labeling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:5157-5163. [PMID: 29577310 DOI: 10.1002/jsfa.9032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND The types and quantity of proteins vary widely between bovine and human milk, with corresponding differences in free and hydrolytic amino acids. In this study, the free and hydrolytic amino acids of bovine and human colostrum were for the first time qualitatively and quantitatively determined using isobaric tags for relative and absolute quantification technology combined with liquid chromatography tandem mass spectrometry detection. RESULTS Total free amino acid content was 0.32 g L-1 and 0.63 g L-1 in bovine and human colostrum respectively, with free amino acid content in human colostrum twice that of bovine colostrum. However, total hydrolytic amino acid content was 4.2 g L-1 and 2.2 g L-1 in bovine and human colostrum respectively. We found that the hydrolytic amino acid content in bovine colostrum was higher than that in human colostrum; however, the amount of free amino acids and the overall amino acid content in human colostrum were respectively substantially higher and more varied than in bovine colostrum. CONCLUSION Our findings revealed differences between bovine and human colostrum, with these data providing the basis for further research into amino acid metabolomics and infant formula. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaona Liang
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Hongjiao Han
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xue Zhao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xueyan Cao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Mei Yang
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Dongbing Tao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Rina Wu
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xiqing Yue
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| |
Collapse
|
84
|
Kahn S, Liao Y, Du X, Xu W, Li J, Lönnerdal B. Exosomal MicroRNAs in Milk from Mothers Delivering Preterm Infants Survive in Vitro Digestion and Are Taken Up by Human Intestinal Cells. Mol Nutr Food Res 2018; 62:e1701050. [PMID: 29644801 DOI: 10.1002/mnfr.201701050] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/08/2018] [Indexed: 01/19/2023]
Abstract
SCOPE This study investigates the ability of preterm milk exosomes to survive gastric/pancreatic digestion, internalization by intestinal epithelia, and the microRNAs (miRNAs) contents. METHODS AND RESULTS At average infant age 1 week and 6 days, milk is collected from mothers who delivered preterm and term infants (n = 10). Milk is exposed to conditions simulating infant gut digestion. Exosomes are isolated and lysed, and the exposed miRNAs are sequenced. Preterm milk exosomes survive in vitro digestion, and can be taken up by intestinal epithelia. Three hundred and thirty miRNAs are identified as preterm milk exosome miRNAs, and in vitro digestion does not have a pronounced effect on their expression. The abundant miRNAs in preterm milk exosomes are similar to those from term milk. Twenty-one low abundance miRNAs are specifically expressed in preterm milk exosomes compared to early term milk in the current study and what previously is found in mature term milk. CONCLUSION These results for the first time reveal the survivability of preterm milk exosomes following simulated gastric/pancreatic digestion. The authors demonstrate the richness of the miRNAs content in these exosomes. The results improve the knowledge of preterm milk biology and the molecular basis by which exosome miRNAs may uniquely affect preterm infants during early development.
Collapse
Affiliation(s)
- Sarah Kahn
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Yalin Liao
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, 6150, Australia
| | - Xiaogu Du
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Wei Xu
- School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA, 6150, Australia
| | - Jie Li
- Genome Centre, University of California, Davis, CA, 95616, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| |
Collapse
|
85
|
de la Torre Gomez C, Goreham RV, Bech Serra JJ, Nann T, Kussmann M. "Exosomics"-A Review of Biophysics, Biology and Biochemistry of Exosomes With a Focus on Human Breast Milk. Front Genet 2018; 9:92. [PMID: 29636770 PMCID: PMC5881086 DOI: 10.3389/fgene.2018.00092] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/05/2018] [Indexed: 12/13/2022] Open
Abstract
Exosomes are biomolecular nanostructures released from cells. They carry specific biomolecular information and are mainly researched for their exquisite properties as a biomarker source and delivery system. We introduce exosomes in the context of other extracellular vesicles, describe their biophysical isolation and characterisation and discuss their biochemical profiling. Motivated by our interest in early-life nutrition and health, and corresponding studies enrolling lactating mothers and their infants, we zoom into exosomes derived from human breast milk. We argue that these should be more extensively studied at proteomic and micronutrient profiling level, because breast milk exosomes provide a more specific window into breast milk quality from an immunological (proteomics) and nutritional (micronutrient) perspective. Such enhanced breast milk exosome profiling would thereby complement and enrich the more classical whole breast milk analysis and is expected to deliver more functional insights than the rather descriptive analysis of human milk, or larger fractions thereof, such as milk fat globule membrane. We substantiate our arguments by a bioinformatic analysis of two published proteomic data sets of human breast milk exosomes.
Collapse
Affiliation(s)
| | - Renee V. Goreham
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington, New Zealand
| | - Joan J. Bech Serra
- Proteomics Unit, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Thomas Nann
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, Wellington, New Zealand
| | - Martin Kussmann
- Liggins Institute, University of Auckland, Auckland, New Zealand
- National Science Challenge “High-Value Nutrition”, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
86
|
Le Doare K, Holder B, Bassett A, Pannaraj PS. Mother's Milk: A Purposeful Contribution to the Development of the Infant Microbiota and Immunity. Front Immunol 2018; 9:361. [PMID: 29599768 PMCID: PMC5863526 DOI: 10.3389/fimmu.2018.00361] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 02/08/2018] [Indexed: 12/12/2022] Open
Abstract
Breast milk is the perfect nutrition for infants, a result of millions of years of evolution. In addition to providing a source of nutrition, breast milk contains a diverse array of microbiota and myriad biologically active components that are thought to guide the infant’s developing mucosal immune system. It is believed that bacteria from the mother’s intestine may translocate to breast milk and dynamically transfer to the infant. Such interplay between mother and her infant is a key to establishing a healthy infant intestinal microbiome. These intestinal bacteria protect against many respiratory and diarrheal illnesses, but are subject to environmental stresses such as antibiotic use. Orchestrating the development of the microbiota are the human milk oligosaccharides (HMOs), the synthesis of which are partially determined by the maternal genotype. HMOs are thought to play a role in preventing pathogenic bacterial adhesion though multiple mechanisms, while also providing nutrition for the microbiome. Extracellular vesicles (EVs), including exosomes, carry a diverse cargo, including mRNA, miRNA, and cytosolic and membrane-bound proteins, and are readily detectable in human breast milk. Strongly implicated in cell–cell signaling, EVs could therefore may play a further role in the development of the infant microbiome. This review considers the emerging role of breast milk microbiota, bioactive HMOs, and EVs in the establishment of the neonatal microbiome and the consequent potential for modulation of neonatal immune system development.
Collapse
Affiliation(s)
- Kirsty Le Doare
- Centre for International Child Health, Imperial College London, London, United Kingdom.,Paediatrics, Imperial College London, London, United Kingdom.,Paediatric Infectious Diseases Research Group, St. George's, University of London, London, United Kingdom.,Vaccines & Immunity Theme, MRC Unit The Gambia, Fajara, Gambia
| | - Beth Holder
- Centre for International Child Health, Imperial College London, London, United Kingdom.,Paediatrics, Imperial College London, London, United Kingdom
| | - Aisha Bassett
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States
| | - Pia S Pannaraj
- Division of Infectious Diseases, Children's Hospital Los Angeles, Los Angeles, CA, United States.,Department of Pediatrics and Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
87
|
Di Venere M, Viglio S, Cagnone M, Bardoni A, Salvini R, Iadarola P. Advances in the analysis of “less-conventional” human body fluids: An overview of the CE- and HPLC-MS applications in the years 2015-2017. Electrophoresis 2017; 39:160-178. [DOI: 10.1002/elps.201700276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/28/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Monica Di Venere
- Department of Molecular Medicine; Biochemistry Unit; University of Pavia; Pavia PV Italy
| | - Simona Viglio
- Department of Molecular Medicine; Biochemistry Unit; University of Pavia; Pavia PV Italy
| | - Maddalena Cagnone
- Department of Molecular Medicine; Biochemistry Unit; University of Pavia; Pavia PV Italy
| | - Anna Bardoni
- Department of Molecular Medicine; Biochemistry Unit; University of Pavia; Pavia PV Italy
| | - Roberta Salvini
- Department of Molecular Medicine; Biochemistry Unit; University of Pavia; Pavia PV Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnologies “L. Spallanzani”; Biochemistry Unit; University of Pavia; Pavia PV Italy
| |
Collapse
|
88
|
Bardanzellu F, Fanos V, Reali A. "Omics" in Human Colostrum and Mature Milk: Looking to Old Data with New Eyes. Nutrients 2017; 9:E843. [PMID: 28783113 PMCID: PMC5579636 DOI: 10.3390/nu9080843] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/21/2022] Open
Abstract
Human Milk (HM) is the best source for newborn nutrition until at least six months; it exerts anti-inflammatory and anti-infective functions, promotes immune system formation and supports organ development. Breastfeeding could also protect from obesity, diabetes and cardiovascular disease. Furthermore, human colostrum (HC) presents a peculiar role in newborn support as a protective effect against allergic and chronic diseases, in addition to long-term metabolic benefits. In this review, we discuss the recent literature regarding "omics" technologies and growth factors (GF) in HC and the effects of pasteurization on its composition. Our aim was to provide new evidence in terms of transcriptomics, proteomics, metabolomics, and microbiomics, also in relation to maternal metabolic diseases and/or fetal anomalies and to underline the functions of GF. Since HC results are so precious, particularly for the vulnerable pre-terms category, we also discuss the importance of HM pasteurization to ensure donated HC even to neonates whose mothers are unable to provide. To the best of our knowledge, this is the first review analyzing in detail the molecular pattern, microbiota, bioactive factors, and dynamic profile of HC, finding clinical correlations of such mediators with their possible in vivo effects and with the consequent impact on neonatal outcomes.
Collapse
Affiliation(s)
- Flaminia Bardanzellu
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, AOU and University of Cagliari, 09124 Cagliari, Italy.
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, AOU and University of Cagliari, 09124 Cagliari, Italy.
| | - Alessandra Reali
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, AOU and University of Cagliari, 09124 Cagliari, Italy.
| |
Collapse
|