151
|
Kupsco A, Prada D, Valvi D, Hu L, Petersen MS, Coull B, Grandjean P, Weihe P, Baccarelli AA. Human milk extracellular vesicle miRNA expression and associations with maternal characteristics in a population-based cohort from the Faroe Islands. Sci Rep 2021; 11:5840. [PMID: 33712635 PMCID: PMC7970999 DOI: 10.1038/s41598-021-84809-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
Human milk plays a critical role in infant development and health, particularly in cognitive, immune, and cardiometabolic functions. Milk contains extracellular vesicles (EVs) that can transport biologically relevant cargo from mother to infant, including microRNAs (miRNAs). We aimed to characterize milk EV-miRNA profiles in a human population cohort, assess potential pathways and ontology, and investigate associations with maternal characteristics. We conducted the first study to describe the EV miRNA profile of human milk in 364 mothers from a population-based mother-infant cohort in the Faroe Islands using small RNA sequencing. We detected 1523 miRNAs with ≥ one read in 70% of samples. Using hierarchical clustering, we determined five EV-miRNA clusters, the top three consisting of 15, 27 and 67 miRNAs. Correlation coefficients indicated that the expression of many miRNAs within the top three clusters was highly correlated. Top-cluster human milk EV-miRNAs were involved in pathways enriched for the endocrine system, cellular community, neurodevelopment, and cancers. miRNA expression was associated with time to milk collection post-delivery, maternal body mass index, and maternal smoking, but not maternal parity. Future studies investigating determinants of human EV-miRNAs and associated health outcomes are needed to elucidate the role of human milk EV-miRNAs in health and disease.
Collapse
Affiliation(s)
- Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10023, USA.
| | - Diddier Prada
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10023, USA
- Unit for Biomedical Research in Cancer, Instituto Nacional de Cancerologia, Universidad Nacional Autonoma de Mexico, 14080, Mexico City, Mexico
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lisa Hu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10023, USA
| | - Maria Skaalum Petersen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Brent Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Medicine, University of Southern Denmark, Odense C, Denmark
| | - Pal Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands
- Center of Health Science, University of the Faroe Islands, Tórshavn, Faroe Islands
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, 10023, USA
| |
Collapse
|
152
|
Zonneveld MI, van Herwijnen MJC, Fernandez-Gutierrez MM, Giovanazzi A, de Groot AM, Kleinjan M, van Capel TMM, Sijts AJAM, Taams LS, Garssen J, de Jong EC, Kleerebezem M, Nolte-'t Hoen ENM, Redegeld FA, Wauben MHM. Human milk extracellular vesicles target nodes in interconnected signalling pathways that enhance oral epithelial barrier function and dampen immune responses. J Extracell Vesicles 2021; 10:e12071. [PMID: 33732416 PMCID: PMC7944547 DOI: 10.1002/jev2.12071] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/18/2020] [Accepted: 01/31/2021] [Indexed: 12/24/2022] Open
Abstract
Maternal milk is nature's first functional food. It plays a crucial role in the development of the infant's gastrointestinal (GI) tract and the immune system. Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer enclosed vesicles released by cells for intercellular communication and are a component of milk. Recently, we discovered that human milk EVs contain a unique proteome compared to other milk components. Here, we show that physiological concentrations of milk EVs support epithelial barrier function by increasing cell migration via the p38 MAPK pathway. Additionally, milk EVs inhibit agonist‐induced activation of endosomal Toll like receptors TLR3 and TLR9. Furthermore, milk EVs directly inhibit activation of CD4+ T cells by temporarily suppressing T cell activation without inducing tolerance. We show that milk EV proteins target key hotspots of signalling networks that can modulate cellular processes in various cell types of the GI tract.
Collapse
Affiliation(s)
- Marijke I Zonneveld
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands.,Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Martijn J C van Herwijnen
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | | | - Alberta Giovanazzi
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Anne Marit de Groot
- Division of Infectious Diseases & Immunology Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Marije Kleinjan
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Toni M M van Capel
- Department of Experimental Immunology Academic Medical Center Amsterdam The Netherlands Centre for inflammation University of Amsterdam Amsterdam Infection & Immunity Institute (AI&II) Amsterdam The Netherlands
| | - Alice J A M Sijts
- Division of Infectious Diseases & Immunology Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Leonie S Taams
- Centre for Inflammation Biology and Cancer Immunology Department of Inflammation Biology School of Immunology & Microbial Sciences King's College London London UK
| | - Johan Garssen
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands.,Global Centre of Excellence Immunology Danone Nutricia Research Utrecht The Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology Academic Medical Center Amsterdam The Netherlands Centre for inflammation University of Amsterdam Amsterdam Infection & Immunity Institute (AI&II) Amsterdam The Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group Department of Animal Sciences Wageningen University Wageningen The Netherlands
| | - Esther N M Nolte-'t Hoen
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| | - Frank A Redegeld
- Division of Pharmacology Department of Pharmaceutical Sciences Faculty of Science Utrecht University Utrecht The Netherlands
| | - Marca H M Wauben
- Department of Biomolecular Health Sciences Faculty of Veterinary Medicine Utrecht University Utrecht The Netherlands
| |
Collapse
|
153
|
Carr LE, Virmani MD, Rosa F, Munblit D, Matazel KS, Elolimy AA, Yeruva L. Role of Human Milk Bioactives on Infants' Gut and Immune Health. Front Immunol 2021; 12:604080. [PMID: 33643310 PMCID: PMC7909314 DOI: 10.3389/fimmu.2021.604080] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 12/26/2022] Open
Abstract
Exclusive human milk feeding of the newborn is recommended during the first 6 months of life to promote optimal health outcomes during early life and beyond. Human milk contains a variety of bioactive factors such as hormones, cytokines, leukocytes, immunoglobulins, lactoferrin, lysozyme, stem cells, human milk oligosaccharides (HMOs), microbiota, and microRNAs. Recent findings highlighted the potential importance of adding HMOs into infant formula for their roles in enhancing host defense mechanisms in neonates. Therefore, understanding the roles of human milk bioactive factors on immune function is critical to build the scientific evidence base around breastfeeding recommendations, and to enhance positive health outcomes in formula fed infants through modifications to formulas. However, there are still knowledge gaps concerning the roles of different milk components, the interactions between the different components, and the mechanisms behind health outcomes are poorly understood. This review aims to show the current knowledge about HMOs, milk microbiota, immunoglobulins, lactoferrin, and milk microRNAs (miRNAs) and how these could have similar mechanisms of regulating gut and microbiota function. It will also highlight the knowledge gaps for future research.
Collapse
Affiliation(s)
- Laura E. Carr
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Misty D. Virmani
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Fernanda Rosa
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Daniel Munblit
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- Inflammation, Repair and Development Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, United Kingdom
- Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| | | | - Ahmed A. Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children's Nutrition Center, Little Rock, AR, United States
- Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
154
|
The Gut‒Breast Axis: Programming Health for Life. Nutrients 2021; 13:nu13020606. [PMID: 33673254 PMCID: PMC7917897 DOI: 10.3390/nu13020606] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 12/16/2022] Open
Abstract
The gut is a pivotal organ in health and disease. The events that take place in the gut during early life contribute to the programming, shaping and tuning of distant organs, having lifelong consequences. In this context, the maternal gut plays a quintessence in programming the mammary gland to face the nutritional, microbiological, immunological, and neuroendocrine requirements of the growing infant. Subsequently, human colostrum and milk provides the infant with an impressive array of nutrients and bioactive components, including microbes, immune cells, and stem cells. Therefore, the axis linking the maternal gut, the breast, and the infant gut seems crucial for a correct infant growth and development. The aim of this article is not to perform a systematic review of the human milk components but to provide an insight of their extremely complex interactions, which render human milk a unique functional food and explain why this biological fluid still truly remains as a scientific enigma.
Collapse
|
155
|
O'Reilly D, Dorodnykh D, Avdeenko NV, Nekliudov NA, Garssen J, Elolimy AA, Petrou L, Simpson MR, Yeruva L, Munblit D. Reply to J Zempleni. Adv Nutr 2021; 12:281. [PMID: 33517406 PMCID: PMC7850015 DOI: 10.1093/advances/nmaa149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
| | - Denis Dorodnykh
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nina V Avdeenko
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nikita A Nekliudov
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ahmed A Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Loukia Petrou
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | | | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | | |
Collapse
|
156
|
The neglected nutrigenomics of milk: What is the role of inter-species transfer of small non-coding RNA? FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
157
|
Wang L, Wang X, Shi Z, Shen L, Zhang J, Zhang J. Bovine milk exosomes attenuate the alteration of purine metabolism and energy status in IEC-6 cells induced by hydrogen peroxide. Food Chem 2021; 350:129142. [PMID: 33610842 DOI: 10.1016/j.foodchem.2021.129142] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 12/13/2022]
Abstract
Evidence suggests that dietary depletion of bovine milk exosomes and their cargos causes a loss of circulating microRNAs and a series of health problems. The aim of the current study was to determine whether bovine milk exosomes affect purine nucleotide metabolism and energy metabolism in oxidatively stressed intestinal crypt epithelial cells (IEC-6). Cells were pretreated with exosomes, followed by H2O2 to induce oxidative stress. Reactive oxidative species (ROS) levels, purine nucleotides, purine metabolic key enzyme activities, cell energy status, and AMPK protein expression were analysed. Exosome pretreatment reduced ROS level and the activities of adenosine deaminase and xanthine oxidase induced by H2O2 in cells. Total adenine nucleotides and energy charge were increased with exosome pretreatment, while the AMPK phosphorylation level was downregulated. The results indicated that bovine milk exosomes could attenuate purine nucleotide catabolism and improve energy status in oxidatively stressed IEC-6 cells and exerted protective effects against oxidative stress.
Collapse
Affiliation(s)
- Lanfang Wang
- Research Center for Translational Medicine at Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, PR China
| | - Xinyan Wang
- The People's Hospital of Zhaoyuan City, Shandong Province, PR China
| | - Zhexi Shi
- Research Center for Translational Medicine at Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, PR China
| | - Li Shen
- Research Center for Translational Medicine at Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, PR China
| | - Jing Zhang
- Research Center for Translational Medicine at Shanghai East Hospital, School of Life Science and Technology, Tongji University, Shanghai, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, PR China.
| | - Jun Zhang
- Research Center for Translational Medicine at Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai, PR China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, PR China.
| |
Collapse
|
158
|
Askenase PW. Ancient Evolutionary Origin and Properties of Universally Produced Natural Exosomes Contribute to Their Therapeutic Superiority Compared to Artificial Nanoparticles. Int J Mol Sci 2021; 22:1429. [PMID: 33572657 PMCID: PMC7866973 DOI: 10.3390/ijms22031429] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Extracellular vesicles (EVs), such as exosomes, are newly recognized fundamental, universally produced natural nanoparticles of life that are seemingly involved in all biologic processes and clinical diseases. Due to their universal involvements, understanding the nature and also the potential therapeutic uses of these nanovesicles requires innovative experimental approaches in virtually every field. Of the EV group, exosome nanovesicles and larger companion micro vesicles can mediate completely new biologic and clinical processes dependent on the intercellular transfer of proteins and most importantly selected RNAs, particularly miRNAs between donor and targeted cells to elicit epigenetic alterations inducing functional cellular changes. These recipient acceptor cells are nearby (paracrine transfers) or far away after distribution via the circulation (endocrine transfers). The major properties of such vesicles seem to have been conserved over eons, suggesting that they may have ancient evolutionary origins arising perhaps even before cells in the primordial soup from which life evolved. Their potential ancient evolutionary attributes may be responsible for the ability of some modern-day exosomes to withstand unusually harsh conditions, perhaps due to unique membrane lipid compositions. This is exemplified by ability of the maternal milk exosomes to survive passing the neonatal acid/enzyme rich stomach. It is postulated that this resistance also applies to their durable presence in phagolysosomes, thus suggesting a unique intracellular release of their contained miRNAs. A major discussed issue is the generally poorly realized superiority of these naturally evolved nanovesicles for therapies when compared to human-engineered artificial nanoparticles, e.g., for the treatment of diseases like cancers.
Collapse
Affiliation(s)
- Phillip W Askenase
- Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
159
|
Melnik BC. Synergistic Effects of Milk-Derived Exosomes and Galactose on α-Synuclein Pathology in Parkinson's Disease and Type 2 Diabetes Mellitus. Int J Mol Sci 2021; 22:1059. [PMID: 33494388 PMCID: PMC7865729 DOI: 10.3390/ijms22031059] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies associate milk consumption with an increased risk of Parkinson's disease (PD) and type 2 diabetes mellitus (T2D). PD is an α-synucleinopathy associated with mitochondrial dysfunction, oxidative stress, deficient lysosomal clearance of α-synuclein (α-syn) and aggregation of misfolded α-syn. In T2D, α-syn promotes co-aggregation with islet amyloid polypeptide in pancreatic β-cells. Prion-like vagal nerve-mediated propagation of exosomal α-syn from the gut to the brain and pancreatic islets apparently link both pathologies. Exosomes are critical transmitters of α-syn from cell to cell especially under conditions of compromised autophagy. This review provides translational evidence that milk exosomes (MEX) disturb α-syn homeostasis. MEX are taken up by intestinal epithelial cells and accumulate in the brain after oral administration to mice. The potential uptake of MEX miRNA-148a and miRNA-21 by enteroendocrine cells in the gut, dopaminergic neurons in substantia nigra and pancreatic β-cells may enhance miRNA-148a/DNMT1-dependent overexpression of α-syn and impair miRNA-148a/PPARGC1A- and miRNA-21/LAMP2A-dependent autophagy driving both diseases. MiRNA-148a- and galactose-induced mitochondrial oxidative stress activate c-Abl-mediated aggregation of α-syn which is exported by exosome release. Via the vagal nerve and/or systemic exosomes, toxic α-syn may spread to dopaminergic neurons and pancreatic β-cells linking the pathogenesis of PD and T2D.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| |
Collapse
|
160
|
González MI, González-Arjona M, Santos-Coquillat A, Vaquero J, Vázquez-Ogando E, de Molina A, Peinado H, Desco M, Salinas B. Covalently Labeled Fluorescent Exosomes for In Vitro and In Vivo Applications. Biomedicines 2021; 9:biomedicines9010081. [PMID: 33467033 PMCID: PMC7829962 DOI: 10.3390/biomedicines9010081] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/31/2020] [Accepted: 01/11/2021] [Indexed: 01/05/2023] Open
Abstract
The vertiginous increase in the use of extracellular vesicles and especially exosomes for therapeutic applications highlights the necessity of advanced techniques for gaining a deeper knowledge of their pharmacological properties. Herein, we report a novel chemical approach for the robust attachment of commercial fluorescent dyes to the exosome surface with covalent binding. The applicability of the methodology was tested on milk and cancer cell-derived exosomes (from U87 and B16F10 cancer cells). We demonstrated that fluorescent labeling did not modify the original physicochemical properties of exosomes. We tested this nanoprobe in cell cultures and healthy mice to validate its use for in vitro and in vivo applications. We confirmed that these fluorescently labeled exosomes could be successfully visualized with optical imaging.
Collapse
Affiliation(s)
- María Isabel González
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (M.I.G.); (M.G.-A.); (A.S.-C.)
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Mario González-Arjona
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (M.I.G.); (M.G.-A.); (A.S.-C.)
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Ana Santos-Coquillat
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (M.I.G.); (M.G.-A.); (A.S.-C.)
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Javier Vaquero
- HepatoGastro Lab, Servicio de Ap. Digestivo del HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (J.V.); (E.V.-O.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
| | - Elena Vázquez-Ogando
- HepatoGastro Lab, Servicio de Ap. Digestivo del HGU Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (J.V.); (E.V.-O.)
| | - Antonio de Molina
- Comparative Medicine Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain;
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Department of Molecular Oncology, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain;
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (M.I.G.); (M.G.-A.); (A.S.-C.)
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
- Correspondence: (M.D.); (B.S.)
| | - Beatriz Salinas
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain; (M.I.G.); (M.G.-A.); (A.S.-C.)
- Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
- Correspondence: (M.D.); (B.S.)
| |
Collapse
|
161
|
Aminzadeh MA, Fournier M, Akhmerov A, Jones‐Ungerleider KC, Valle JB, Marbán E. Casein-enhanced uptake and disease-modifying bioactivity of ingested extracellular vesicles. J Extracell Vesicles 2021; 10:e12045. [PMID: 33456725 PMCID: PMC7798403 DOI: 10.1002/jev2.12045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/02/2020] [Accepted: 12/01/2020] [Indexed: 01/28/2023] Open
Abstract
Extracellular vesicles (EVs) from cardiac stromal cells, developed as therapeutic candidates, improve dystrophic muscle function when administered parenterally, but oral delivery remains untested. We find that casein, the dominant protein in breast milk, enhances the uptake and bioactivity of ingested heart-derived EVs, altering gene expression in blood cells and enhancing muscle function in mdx mice with muscular dystrophy. Thus, EVs, administered orally, are absorbed and exert disease-modifying bioactivity in vivo. Formulating EVs with casein enhances uptake and markedly expands the range of potential therapeutic applications.
Collapse
Affiliation(s)
- Mark A. Aminzadeh
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCaliforniaUSA
| | - Mario Fournier
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCaliforniaUSA
| | | | | | - Jackelyn B. Valle
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCaliforniaUSA
| | - Eduardo Marbán
- Cedars‐Sinai Medical CenterSmidt Heart InstituteLos AngelesCaliforniaUSA
| |
Collapse
|
162
|
Ma S, Niu M, Hao Z, Liu M, Tong C, Zhao X. Selective packaged circular RNAs in milk extracellular vesicles during Staphylococcus aureus infection may have potential against bacterial infection. RNA Biol 2020; 18:818-831. [PMID: 33241726 DOI: 10.1080/15476286.2020.1853975] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Extracellular vesicles (EVs) provide a novel intercellular communication mechanism to transfer biologically important molecules to target cells. Although several pieces of evidence have shown that EVs have potential to respond to bacterial infections, our knowledge about the role of circular RNA (circRNA), an important cargo of EV, behind this process remains poor. In particular, the mechanism by which circRNAs are packaged into EVs remains elusive during bacterial infection. In the present study, EVs from bovine milk samples with or without Staphylococcus aureus (S. aureus) infection were isolated. The presence of circRNAs in milk-derived EVs (MEVs) was validated for the first time by PCR amplification with convergent and divergent primers and the RNase R resistance test. Through high-throughput sequencing, the expression profile of circRNAs in EVs was found to be changed during S. aureus infection. Moreover, we demonstrated that circRNAs were selectively packaged into EVs. Finally, bioinformatic analyses predicted the involvement of differentially expressed circRNAs in immune functions. In summary, our findings offer an insight into the packaging mechanism of EV circRNAs and underscore the potential by which host used the EV circRNAs in response to pathogenic bacterial infections.
Collapse
Affiliation(s)
- Shaoyang Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingze Niu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zehua Hao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Miaomiao Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Tong
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xin Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.,Department of Animal Science, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
163
|
Pelissier Vatter FA, Lucotti S, Zhang H. Recent Advances in Experimental Models of Breast Cancer Exosome Secretion, Characterization and Function. J Mammary Gland Biol Neoplasia 2020; 25:305-317. [PMID: 33351162 DOI: 10.1007/s10911-020-09473-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is responsible for 15% of all the cancer deaths among women in the USA. The tumor microenvironment (TME) has the potential to act as a driver of breast cancer progression and metastasis. The TME is composed of stromal cells within an extracellular matrix and soluble cytokines, chemokines and extracellular vesicles and nanoparticles that actively influence cell behavior. Extracellular vesicles include exosomes, microvesicles and large oncosomes that orchestrate fundamental processes during tumor progression through direct interaction with target cells. Long before tumor cell spread to future metastatic sites, tumor-secreted exosomes enter the circulation and establish distant pre-metastatic niches, hospitable and permissive milieus for metastatic colonization. Emerging evidence suggests that breast cancer exosomes promote tumor progression and metastasis by inducing vascular leakiness, angiogenesis, invasion, immunomodulation and chemoresistance. Exosomes are found in almost all physiological fluids including plasma, urine, saliva, and breast milk, providing a valuable resource for the development of non-invasive cancer biomarkers. Here, we review work on the role of exosomes in breast cancer progression and metastasis, and describe the most recent advances in models of exosome secretion, isolation, characterization and functional analysis. We highlight the potential applications of plasma-derived exosomes as predictive biomarkers for breast cancer diagnosis, prognosis and therapy monitoring. We finally describe the therapeutic approaches of exosomes in breast cancer.
Collapse
Affiliation(s)
- Fanny A Pelissier Vatter
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA.
| | - Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - Haiying Zhang
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
164
|
Grossen P, Portmann M, Koller E, Duschmalé M, Minz T, Sewing S, Pandya NJ, van Geijtenbeek SK, Ducret A, Kusznir EA, Huber S, Berrera M, Lauer ME, Ringler P, Nordbo B, Jensen ML, Sladojevich F, Jagasia R, Alex R, Gamboni R, Keller M. Evaluation of bovine milk extracellular vesicles for the delivery of locked nucleic acid antisense oligonucleotides. Eur J Pharm Biopharm 2020; 158:198-210. [PMID: 33248268 DOI: 10.1016/j.ejpb.2020.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Abstract
The natural capacity of extracellular vesicles (EVs) to transport their payload to recipient cells has raised big interest to repurpose EVs as delivery vehicles for xenobiotics. In the present study, bovine milk-derived EVs (BMEVs) were investigated for their potential to shuttle locked nucleic acid-modified antisense oligonucleotides (LNA ASOs) into the systemic circulation after oral administration. To this end, a broad array of analytical methods including proteomics and lipidomics were used to thoroughly characterize BMEVs. We found that additional purification by density gradients efficiently reduced levels of non-EV associated proteins. The potential of BMEVs to functionally transfer LNA ASOs was tested using advanced in vitro systems (i.e. hPSC-derived neurons and primary human cells). A slight increase in cellular LNA ASO internalization and target gene reduction was observed when LNA ASOs were delivered using BMEVs. When dosed orally in mice, only a small fraction (about 1% of total administered dose) of LNA ASOs was recovered in the peripheral tissues liver and kidney, however, no significant reduction in target gene expression (i.e. functional knockdown) was observed.
Collapse
Affiliation(s)
- Philip Grossen
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Michaela Portmann
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Erich Koller
- Roche Pharma Research and Early Development, DMPK, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Martina Duschmalé
- Roche Pharma Research and Early Development, iSafe, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Tanja Minz
- Roche Pharma Research and Early Development, iSafe, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Sabine Sewing
- Roche Pharma Research and Early Development, iSafe, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Nikhil Janak Pandya
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland; Roche Pharma Research and Early Development, Neurology and Rare Diseases Disease Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Sabine Kux van Geijtenbeek
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Axel Ducret
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Eric-André Kusznir
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Sylwia Huber
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Marco Berrera
- Roche Pharma Research and Early Development, Biomics and Pathology, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Matthias E Lauer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Philippe Ringler
- Center for Cellular Imaging and NanoAnalytics (C-CINA), University of Basel, Basel, Switzerland
| | - Bettina Nordbo
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd, Fremtidsvej3, 2970 Hoersholm, Denmark
| | - Marianne Lerbech Jensen
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Copenhagen, F. Hoffmann-La Roche Ltd, Fremtidsvej3, 2970 Hoersholm, Denmark
| | - Filippo Sladojevich
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Ravi Jagasia
- Roche Pharma Research and Early Development, Neurology and Rare Diseases Disease Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Rainer Alex
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Remo Gamboni
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Michael Keller
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070 Basel, Switzerland.
| |
Collapse
|
165
|
Gao HN, Ren FZ, Wen PC, Xie LX, Wang R, Yang ZN, Li YX. Yak milk-derived exosomal microRNAs regulate intestinal epithelial cells on proliferation in hypoxic environment. J Dairy Sci 2020; 104:1291-1303. [PMID: 33246613 DOI: 10.3168/jds.2020-19063] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/08/2020] [Indexed: 01/07/2023]
Abstract
Intestinal epithelial cells (IEC) act as an important intestinal barrier whose function can be impaired upon induction by hypoxia. Although intestinal barrier injuries are preventable by milk-derived exosomal microRNAs (miRNAs), the underlying mechanism remains poorly understood. This study aimed to characterize the effect of yak and cow milk-derived exosomal miRNA on the barrier function of IEC-6 under hypoxic conditions, and explore the mechanism of yak milk exosomal miRNA to relieve the hypoxia stress. First, by Illumina HiSeq 2500 (Illumina Inc., San Diego, CA) sequencing, the miRNA expression was systematically screened, and differential expression of 130 miRNAs was identified with 51 being upregulated and 79 downregulated in yak and cow milk-derived exosomes. Furthermore, the top 20 miRNAs that had a relatively consistent high expression in yak milk exosome were identified, and bta-miR-34a was found to be an effective regulator for alleviating hypoxic injury of IEC-6. In vitro assay of the role of bta-miR-34a on survival of IEC-6 in hypoxia by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) confirmed its effectiveness to significantly increase IEC-6 survival up to 13% for 12 h, and up to 9.5% for 24 h. Investigation on the regulatory relationship between bta-miRNA-34a and the hypoxia-inducible factor/apoptosis signaling pathway provided insights into the possible mechanisms by which bta-miR-34a activated the hypoxia-inducible factor and apoptosis signaling pathway, thus promoting IEC-6 survival. The results of this study suggest an important relationship between miRNA expression and intestine barrier integrity, which facilitated further understanding of the physiological function of yak and cow milk exosomal miRNAs, as well as mechanisms of hypoxia-driven epithelial homeostasis.
Collapse
Affiliation(s)
- H N Gao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - F Z Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - P C Wen
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou 730070, China
| | - L X Xie
- Treasure of Tibet Yak Dairy Co. Ltd., Lhasa, 610000, China
| | - R Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Z N Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China.
| | - Y X Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
166
|
The Physiological MicroRNA Landscape in Nipple Aspirate Fluid: Differences and Similarities with Breast Tissue, Breast Milk, Plasma and Serum. Int J Mol Sci 2020; 21:ijms21228466. [PMID: 33187146 PMCID: PMC7696615 DOI: 10.3390/ijms21228466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Background: MicroRNAs (miRNAs) target 60% of human messenger RNAs and can be detected in tissues and biofluids without loss of stability during sample processing, making them highly appraised upcoming biomarkers for evaluation of disease. However, reporting of the abundantly expressed miRNAs in healthy samples is often surpassed. Here, we characterized for the first time the physiological miRNA landscape in a biofluid of the healthy breast: nipple aspirate fluid (NAF), and compared NAF miRNA expression patterns with publically available miRNA expression profiles of healthy breast tissue, breast milk, plasma and serum. Methods: MiRNA RT-qPCR profiling of NAF (n = 41) and serum (n = 23) samples from two healthy female cohorts was performed using the TaqMan OpenArray Human Advanced MicroRNA 754-Panel. MiRNA quantification data based on non-targeted or multi-targeted profiling techniques for breast tissue, breast milk, plasma and serum were retrieved from the literature by means of a systematic search. MiRNAs from each individual study were orderly ranked between 1 and 50, combined into an overall ranking per sample type and compared. Results: NAF expressed 11 unique miRNAs and shared 21/50 miRNAs with breast tissue. Seven miRNAs were shared between the five sample types. Overlap between sample types varied between 42% and 62%. Highly ranked NAF miRNAs have established roles in breast carcinogenesis. Conclusion: This is the first study to characterize and compare the unique physiological NAF-derived miRNA landscape with the physiological expression pattern in breast tissue, breast milk, plasma and serum. Breast-specific sources did not mutually overlap more than with systemic sources. Given their established role in carcinogenesis, NAF miRNA assessment could be a valuable tool in breast tumor diagnostics.
Collapse
|
167
|
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
|
168
|
Zeng B, Chen T, Luo JY, Zhang L, Xi QY, Jiang QY, Sun JJ, Zhang YL. Biological Characteristics and Roles of Noncoding RNAs in Milk-Derived Extracellular Vesicles. Adv Nutr 2020; 12:1006-1019. [PMID: 33080010 PMCID: PMC8166544 DOI: 10.1093/advances/nmaa124] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) have diverse roles in the transport of proteins, lipids, and nucleic acids between cells, and they serve as mediators of intercellular communication. Noncoding RNAs (ncRNAs) that are present in EVs, including microRNAs, long noncoding RNAs, and circular RNAs, have been found to participate in complex networks of interactions and regulate a wide variety of genes in animals. Milk is an important source of nutrition for humans and other mammals. Evidence suggests that milk-derived EVs contain abundant ncRNAs, which are stable and can be transported to the offspring and other consumers. Current data suggest a strong link between milk EV ncRNAs and many biological processes, and these ncRNAs have been drawing increasing attention and might play an epigenetic regulatory role in recipients, though further research is still necessary to understand their precise roles. The present review introduces basic information about milk EV ncRNAs, summarizes their expression profiles, biological characteristics, and functions based on current knowledge, and discusses their biological roles, indeterminate issues, and perspectives. Our goal is to provide a deeper understanding of the physiological effects of milk EV ncRNAs on offspring and to provide a reference for future research in this field.
Collapse
Affiliation(s)
- Bin Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jun-Yi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Lin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qian-Yun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qing-Yan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou, China
| | | | | |
Collapse
|
169
|
Mandala A, Janssen RC, Palle S, Short KR, Friedman JE. Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms. Nutrients 2020; 12:E3166. [PMID: 33081177 PMCID: PMC7602751 DOI: 10.3390/nu12103166] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the number one chronic liver disease worldwide and is estimated to affect nearly 40% of obese youth and up to 10% of the general pediatric population without any obvious signs or symptoms. Although the early stages of NAFLD are reversible with diet and lifestyle modifications, detecting such stages is hindered by a lack of non-invasive methods of risk assessment and diagnosis. This absence of non-invasive means of diagnosis is directly related to the scarcity of long-term prospective studies of pediatric NAFLD in children and adolescents. In the majority of pediatric NAFLD cases, the mechanisms driving the origin and rapid progression of NAFLD remain unknown. The progression from NAFLD to non-alcoholic steatohepatitis (NASH) in youth is associated with unique histological features and possible immune processes and metabolic pathways that may reflect different mechanisms compared with adults. Recent data suggest that circulating microRNAs (miRNAs) are important new biomarkers underlying pathways of liver injury. Several factors may contribute to pediatric NAFLD development, including high-sugar diets, in utero exposures via epigenetic alterations, changes in the neonatal microbiome, and altered immune system development and mitochondrial function. This review focuses on the unique aspects of pediatric NAFLD and how nutritional exposures impact the immune system, mitochondria, and liver/gastrointestinal metabolic health. These factors highlight the need for answers to how NAFLD develops in children and for early stage-specific interventions.
Collapse
Affiliation(s)
- Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Sirish Palle
- Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Kevin R. Short
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
170
|
Carrillo-Lozano E, Sebastián-Valles F, Knott-Torcal C. Circulating microRNAs in Breast Milk and Their Potential Impact on the Infant. Nutrients 2020; 12:E3066. [PMID: 33049923 PMCID: PMC7601398 DOI: 10.3390/nu12103066] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (MiRNAs) are small RNA molecules that can exert regulatory functions in gene expression. MiRNAs have been identified in diverse tissues and biological fluids, both in the context of health and disease. Breastfeeding has been widely recognized for its superior nutritional benefits; however, a number of bioactive compounds have been found to transcend these well-documented nutritional contributions. Breast milk was identified as a rich source of miRNAs. There has been increasing interest about their potential ability to transfer to the offspring as well as what their specific involvement is within the benefits of breast milk in the infant. In comparison to breast milk, formula milk lacks many of the benefits of breastfeeding, which is thought to be a result of the absence of some of these bioactive compounds. In recent years, the miRNA profile of breast milk has been widely studied, along with the possible transfer mechanisms throughout the infant's digestive tract and the role of miRNA-modulated genes and their potential protective and regulatory functions. Nonetheless, to date, the current evidence is not consistent, as many methodological limitations have been identified; hence, discrepancies exits about the biological functions of miRNAs. Further research is needed to provide thorough knowledge in this field.
Collapse
|
171
|
Kim KU, Kim WH, Jeong CH, Yi DY, Min H. More than Nutrition: Therapeutic Potential of Breast Milk-Derived Exosomes in Cancer. Int J Mol Sci 2020; 21:E7327. [PMID: 33023062 PMCID: PMC7582863 DOI: 10.3390/ijms21197327] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022] Open
Abstract
Human breast milk (HBM) is an irreplaceable source of nutrition for early infant growth and development. Breast-fed children are known to have a low prevalence and reduced risk of various diseases, such as necrotizing enterocolitis, gastroenteritis, acute lymphocytic leukemia, and acute myeloid leukemia. In recent years, HBM has been found to contain a microbiome, extracellular vesicles or exosomes, and microRNAs, as well as nutritional components and non-nutritional proteins, including immunoregulatory proteins, hormones, and growth factors. Especially, the milk-derived exosomes exert various physiological and therapeutic function in cell proliferation, inflammation, immunomodulation, and cancer, which are mainly attributed to their cargo molecules such as proteins and microRNAs. The exosomal miRNAs are protected from enzymatic digestion and acidic conditions, and play a critical role in immune regulation and cancer. In addition, the milk-derived exosomes are developed as drug carriers for delivering small molecules and siRNA to tumor sites. In this review, we examined the various components of HBM and their therapeutic potential, in particular of exosomes and microRNAs, towards cancer.
Collapse
Affiliation(s)
- Ki-Uk Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (K.-U.K.); (W.-H.K.); (C.H.J.)
| | - Wan-Hoon Kim
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (K.-U.K.); (W.-H.K.); (C.H.J.)
| | - Chi Hwan Jeong
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (K.-U.K.); (W.-H.K.); (C.H.J.)
| | - Dae Yong Yi
- Department of Pediatrics, Chung-Ang University College of Medicine, Seoul 06974, Korea
- Department of Pediatrics, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, Seoul 06974, Korea; (K.-U.K.); (W.-H.K.); (C.H.J.)
| |
Collapse
|
172
|
Mecocci S, Gevi F, Pietrucci D, Cavinato L, Luly FR, Pascucci L, Petrini S, Ascenzioni F, Zolla L, Chillemi G, Cappelli K. Anti-Inflammatory Potential of Cow, Donkey and Goat Milk Extracellular Vesicles as Revealed by Metabolomic Profile. Nutrients 2020; 12:E2908. [PMID: 32977543 PMCID: PMC7598260 DOI: 10.3390/nu12102908] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, extracellular vesicles (EVs), cell-derived micro and nano-sized structures enclosed in a double-layer membrane, have been in the spotlight for their high potential in diagnostic and therapeutic applications. Indeed, they act as signal mediators between cells and/or tissues through different mechanisms involving their complex cargo and exert a number of biological effects depending upon EVs subtype and cell source. Being produced by almost all cell types, they are found in every biological fluid including milk. Milk EVs (MEVs) can enter the intestinal cells by endocytosis and protect their labile cargos against harsh conditions in the intestinal tract. In this study, we performed a metabolomic analysis of MEVs, from three different species (i.e., bovine, goat and donkey) by mass spectroscopy (MS) coupled with Ultrahigh-performance liquid chromatography (UHPLC). Metabolites, both common or specific of a species, were identified and enriched metabolic pathways were investigated, with the final aim to evaluate their anti-inflammatory and immunomodulatory properties in view of prospective applications as a nutraceutical in inflammatory conditions. In particular, metabolites transported by MEVs are involved in common pathways among the three species. These metabolites, such as arginine, asparagine, glutathione and lysine, show immunomodulating effects. Moreover, MEVs in goat milk showed a greater number of enriched metabolic pathways as compared to the other kinds of milk.
Collapse
Affiliation(s)
- Samanta Mecocci
- Dipartimento di Medicina Veterinaria, University of Perugia, 06123 Perugia, Italy; (S.M.); (L.P.)
- Centro di Ricerca sul Cavallo Sportivo, University of Perugia, 06123 Perugia, Italy
| | - Federica Gevi
- Dipartimento di Scienze Ecologiche e Biologiche, Università della Tuscia, 01100 Viterbo, Italy; (F.G.); (L.Z.)
| | - Daniele Pietrucci
- Dipartimento per l’Innovazione Nei Sistemi Biologici, Agroalimentari e Forestali, Università della Tuscia, 01100 Viterbo, Italy;
| | - Luca Cavinato
- Dipartimento di Biologia e Biotecnologie C. Darwin, Università di Roma la Sapienza, 00185 Roma, Italy; (L.C.); (F.R.L.); (F.A.)
| | - Francesco R. Luly
- Dipartimento di Biologia e Biotecnologie C. Darwin, Università di Roma la Sapienza, 00185 Roma, Italy; (L.C.); (F.R.L.); (F.A.)
| | - Luisa Pascucci
- Dipartimento di Medicina Veterinaria, University of Perugia, 06123 Perugia, Italy; (S.M.); (L.P.)
| | - Stefano Petrini
- Istituto Zooprofilattico Sperimentale dell’Umbria e delle Marche, 06126 Perugia, Italy;
| | - Fiorentina Ascenzioni
- Dipartimento di Biologia e Biotecnologie C. Darwin, Università di Roma la Sapienza, 00185 Roma, Italy; (L.C.); (F.R.L.); (F.A.)
| | - Lello Zolla
- Dipartimento di Scienze Ecologiche e Biologiche, Università della Tuscia, 01100 Viterbo, Italy; (F.G.); (L.Z.)
| | - Giovanni Chillemi
- Dipartimento per l’Innovazione Nei Sistemi Biologici, Agroalimentari e Forestali, Università della Tuscia, 01100 Viterbo, Italy;
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, CNR, 70126 Bari, Italy
| | - Katia Cappelli
- Dipartimento di Medicina Veterinaria, University of Perugia, 06123 Perugia, Italy; (S.M.); (L.P.)
- Centro di Ricerca sul Cavallo Sportivo, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
173
|
Beeraka NM, Doreswamy SH, Sadhu SP, Srinivasan A, Pragada RR, Madhunapantula SV, Aliev G. The Role of Exosomes in Stemness and Neurodegenerative Diseases-Chemoresistant-Cancer Therapeutics and Phytochemicals. Int J Mol Sci 2020; 21:ijms21186818. [PMID: 32957534 PMCID: PMC7555629 DOI: 10.3390/ijms21186818] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/15/2022] Open
Abstract
Exosomes exhibit a wide range of biological properties and functions in the living organisms. They are nanometric vehicles and used for delivering drugs, as they are biocompatible and minimally immunogenic. Exosomal secretions derived from cancer cells contribute to metastasis, immortality, angiogenesis, tissue invasion, stemness and chemo/radio-resistance. Exosome-derived microRNAs (miRNAs) and long non-coding RNAs (lnc RNAs) are involved in the pathophysiology of cancers and neurodegenerative diseases. For instance, exosomes derived from mesenchymal stromal cells, astrocytes, macrophages, and acute myeloid leukemia (AML) cells are involved in the cancer progression and stemness as they induce chemotherapeutic drug resistance in several cancer cells. This review covered the recent research advances in understanding the role of exosomes in cancer progression, metastasis, angiogenesis, stemness and drug resistance by illustrating the modulatory effects of exosomal cargo (ex. miRNA, lncRNAs, etc.) on cell signaling pathways involved in cancer progression and cancer stem cell growth and development. Recent reports have implicated exosomes even in the treatment of several cancers. For instance, exosomes-loaded with novel anti-cancer drugs such as phytochemicals, tumor-targeting proteins, anticancer peptides, nucleic acids are known to interfere with drug resistance pathways in several cancer cell lines. In addition, this review depicted the need to develop exosome-based novel diagnostic biomarkers for early detection of cancers and neurodegenerative disease. Furthermore, the role of exosomes in stroke and oxidative stress-mediated neurodegenerative diseases including Alzheimer’s disease (AD), and Parkinson’s disease (PD) is also discussed in this article.
Collapse
Affiliation(s)
- Narasimha M. Beeraka
- Center of Excellence in Regenerative Medicine and Molecular Biology (CERM), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India; (N.M.B.); (S.H.D.)
| | - Shalini H. Doreswamy
- Center of Excellence in Regenerative Medicine and Molecular Biology (CERM), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India; (N.M.B.); (S.H.D.)
| | - Surya P. Sadhu
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, Andhra Pradesh, India; (S.P.S.); (R.R.P.)
| | - Asha Srinivasan
- Center of Excellence in Regenerative Medicine and Molecular Biology (CERM), Division of Nanoscience and Technology, Faculty of Life Sciences, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India;
| | - Rajeswara Rao Pragada
- AU College of Pharmaceutical Sciences, Andhra University, Visakhapatnam 530003, Andhra Pradesh, India; (S.P.S.); (R.R.P.)
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
- Special Interest Group in Cancer Biology and Cancer Stem Cells (SIG-CBCSC), JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru 570015, Karnataka, India
- Correspondence: (S.V.M.); or (G.A.); Tel.: +1-440-263-7461 or +7-964-493-1515 (G.A.)
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, 142432 Chernogolovka, Moscow Region, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, 117418 Moscow, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
- Correspondence: (S.V.M.); or (G.A.); Tel.: +1-440-263-7461 or +7-964-493-1515 (G.A.)
| |
Collapse
|
174
|
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: 47] [Impact Index Per Article: 9.4] [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
|
175
|
Pawlowski K, Lago-Novais D, Bevilacqua C, Mobuchon L, Crapart N, Faulconnier Y, Boby C, Carvalho G, Martin P, Leroux C. Different miRNA contents between mammary epithelial cells and milk fat globules: a random or a targeted process? Mol Biol Rep 2020; 47:8259-8264. [PMID: 32909217 DOI: 10.1007/s11033-020-05787-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023]
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs present in milk-derived extracellular vesicles and milk fat globules (MFG). Nucleic acid content between the lactating mammary tissue (MT) and MFG are quite similar but discrepancies exist in their miRNA content. Our objective was to identify the origin of these discrepancies, and to evaluate the existence of a possible mechanism sorting miRNAs that will or will not be exported from the mammary epithelial cells (MECs) in bovine MFG. miR-125b-5p, miR-126-3p, miR-141-3p, and miR-204-5p, chosen on the basis of their abundance in the MT, were quantified using RT-qPCR in lactating cow MT, MFG, and laser capture-microdissected MECs. Two miRNAs (miR-125b-5p and miR-141-3p) were detected in the MT as well as in MFG and MECs. miR-204-5p was detected only in the MT, suggesting that it is very likely expressed in a cell type other than MECs. miR-126-3p was detected both in the MT and in MECs but not in MFG, suggesting a targeting mechanism for miRNAs in MECs. These results highlights differences in miRNA content between MECs and MFG may be due to a possibly not random mechanism for loading MFG with miRNA cargos that could involve a variable distribution in MECs or a sorting mechanism.
Collapse
Affiliation(s)
- Karol Pawlowski
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France.,Department of Pathology and Veterinary Diagnostics, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Daiane Lago-Novais
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France.,Programa de Pós Graduação em Zootecnia, Universidade Federal da Bahia (UFBA), Av. Adhemar de Barros 500, CEP 40170-110, Salvador, BA, Brazil
| | - Claudia Bevilacqua
- UMR1313 Génétique Animale et Biologie Intégrative, AgroParisTech, Université Paris-Saclay, INRAE, 78350, Jouy-en-Josas, France.,Plateforme @BRIDGE, INRAE, Domaine de Vilvert, 78350, Jouy-en-Josas, France
| | - Lenha Mobuchon
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France.,UMR1313 Génétique Animale et Biologie Intégrative, AgroParisTech, Université Paris-Saclay, INRAE, 78350, Jouy-en-Josas, France
| | - Nicolas Crapart
- UMR1313 Génétique Animale et Biologie Intégrative, AgroParisTech, Université Paris-Saclay, INRAE, 78350, Jouy-en-Josas, France.,Plateforme @BRIDGE, INRAE, Domaine de Vilvert, 78350, Jouy-en-Josas, France.,Excilone, 78990, Elancourt, France
| | - Yannick Faulconnier
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France
| | - Céline Boby
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France
| | - Gleidson Carvalho
- Programa de Pós Graduação em Zootecnia, Universidade Federal da Bahia (UFBA), Av. Adhemar de Barros 500, CEP 40170-110, Salvador, BA, Brazil
| | - Patrice Martin
- UMR1313 Génétique Animale et Biologie Intégrative, AgroParisTech, Université Paris-Saclay, INRAE, 78350, Jouy-en-Josas, France
| | - Christine Leroux
- UMR1213 Herbivores, INRAE, Université Clermont-Auvergne, VetAgro Sup, 63122, Saint Genès Champanelle, France.
| |
Collapse
|
176
|
O'Reilly D, Dorodnykh D, Avdeenko NV, Nekliudov NA, Garssen J, Elolimy AA, Petrou L, Simpson MR, Yeruva L, Munblit D. Perspective: The Role of Human Breast-Milk Extracellular Vesicles in Child Health and Disease. Adv Nutr 2020; 12:59-70. [PMID: 32838428 PMCID: PMC7849950 DOI: 10.1093/advances/nmaa094] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
Human breast milk (HM) contains multiple bioactive substances determining its impact on children's health. Extracellular vesicles (EVs) are a heterogeneous group of secreted nanoparticles that are present in HM and may be partially responsible for its beneficial effects. The precise roles and content of EVs in HM remain largely unknown. To examine this, we performed a short narrative review on the literature focusing on HM EVs to contextualize the available data, followed by a scoping review of MEDLINE and Embase databases. We identified 424 nonduplicate citations with 19 original studies included. In this perspective, we summarize the evidence around HM EVs, highlight some theoretical considerations based on existing evidence, and provide an overview of some challenges associated with the complexity and heterogeneity of EV research. We consider how the existing data from HM studies conform to the minimal information for studies of EVs (MISEV) guidelines. Across the studies a variety of research methods were utilized involving both bench-based and translational methods, and a range of different EV contents were examined including RNA, proteins, and glycopeptides. We observed a variety of health outcomes in these studies, including allergy and atopy, necrotizing enterocolitis, and HIV. While some promising results have been demonstrated, the heterogeneity in outcomes of interest, methodological limitations, and relatively small number of studies in the field make comparison between studies or further translational work problematic. To date, no studies have examined normative values of HM EVs in a large, diverse population or with respect to potentially important influencing factors such as timing (hind- vs. foremilk), stage (colostrum vs. mature milk), and infant age (preterm vs. term), which makes extrapolation from bench or "basic" research impossible. Future research should focus on addressing the current inadequacies in the literature and utilize MISEV guidelines to inform study design.
Collapse
Affiliation(s)
| | - Denis Dorodnykh
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nina V Avdeenko
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nikita A Nekliudov
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ahmed A Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Loukia Petrou
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway,Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | | |
Collapse
|
177
|
Abstract
Recent advances on milk exosomes (EXO), cargoes in cell-cell communication, explored their role within and between individuals, including in dairy species. The potential use of EXO as biomarkers of disease and metabolic conditions adds significant interest to the study of EXO in milk. Although several researches have been carried out on circulating miRNA in the milk, less information is available about milk-derived exosomal miRNAs, which are stable over time and resistant to digestion and milk processing. EXO are taken up by recipient cells through specific mechanisms, which enable the selective delivery of cargoes. This suggests that EXO cargoes can be used as biomarkers of health. Nevertheless, methodological limitations and potential applications of milk EXO in dairy ruminants must be considered. The paucity of studies that associate the EXO cargo to specific challenges deserves further investigations to unravel the variation of miRNA and proteins cargo in relation to metabolic imbalance and infectious disease of the mammary gland.
Collapse
|
178
|
Zhou Q, Xie F, Zhou B, Li C, Kang Y, Wu B, Li L, Dai R. Fetal bovine serum-derived exosomes regulate the adipogenic differentiation of human bone marrow mesenchymal stromal cells in a cross-species manner. Differentiation 2020; 115:11-21. [PMID: 32771719 DOI: 10.1016/j.diff.2020.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 05/15/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023]
Abstract
Fetal bovine serum (FBS) contains a large number of exosomes which may disturb the analysis of exosomes derived from cultured cells. We investigated the effect of FBS-derived exosomes (FBS-Exos) on the adipogenic differentiation of human bone marrow mesenchymal stromal cells (hBM-MSCs) and the underlying molecular mechanism. The uptake of FBS-Exos by hBM-MSCs could be detected by the laser confocal microscopy, and the treatment of exosomes resulted in the decreased lipid droplet formation and reduced expression of genes associated with adipogenic differentiation of hBM-MSCs. miR-1246 was identified as an abundant microRNA in FBS-Exos by public sequencing data identification and RT-qPCR validation. Moreover, miR-1246 overexpression in hBM-MSCs led to decreased adipogenic differentiation level, while miR-1246 knockdown in FBS-Exos attenuated the inhibitory effect on the adipogenic differentiation. Our results indicate that FBS-Exos inhibit the adipogenic differentiation of hBM-MSCs in a cross-species manner and miR-1246 transferred by FBS-Exos partly contributes to this effect.
Collapse
Affiliation(s)
- Qiongfei Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China
| | - Fen Xie
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China; Department of Endocrinology and Metabolism, Xiangtan Central Hospital, Xiangtan, Hunan, 411100, China
| | - Bin Zhou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Chan Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China
| | - Yijun Kang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Bo Wu
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China
| | - Lin Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China; Department of Endocrinology and Metabolism, The First Hospital of Changsha, Changsha, Hunan, 410005, China
| | - Ruchun Dai
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, 410011, China; Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Changsha, Hunan, 410011, China.
| |
Collapse
|
179
|
Carr LE, Bowlin AK, Elolimy AA, Byrum SD, Washam CL, Randolph CE, MacLeod SL, Yeruva L. Neonatal Diet Impacts Circulatory miRNA Profile in a Porcine Model. Front Immunol 2020; 11:1240. [PMID: 32655560 PMCID: PMC7324749 DOI: 10.3389/fimmu.2020.01240] [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: 03/03/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022] Open
Abstract
microRNAs (miRNAs) are conserved non-coding small nucleotide molecules found in nearly all species and breastmilk. miRNAs present in breastmilk are very stable to freeze-thaw, RNase treatment, and low pH as they are protected inside exosomes. They are involved in regulating several physiologic and pathologic processes, including immunologic pathways, and we have demonstrated better immune response to vaccines in piglets fed with human milk (HM) in comparison to dairy-based formula (MF). To understand if neonatal diet impacts circulatory miRNA expression, serum miRNA expression was evaluated in piglets fed HM or MF while on their neonatal diet at postnatal day (PND) 21 and post-weaning to solid diet at PND 35 and 51. MF fed piglets showed increased expression of 14 miRNAs and decreased expression of 10 miRNAs, relative to HM fed piglets at PND 21. At PND 35, 9 miRNAs were downregulated in the MF compared to the HM group. At PND 51, 10 miRNAs were decreased and 17 were increased in the MF relative to HM suggesting the persistent effect of neonatal diet. miR-148 and miR-181 were decreased in MF compared to HM at PND 21. Let-7 was decreased at PND 35 while miR-199a and miR-199b were increased at PND 51 in MF compared to HM. Pathway analysis suggested that many of the miRNAs are involved in immune function. In conclusion, we observed differential expression of blood miRNAs at both PND 21 and PND 51. miRNA found in breastmilk were decreased in the serum of the MF group, suggesting that diet impacts circulating miRNA profiles at PND 21. The miRNAs continue to be altered at PND 51 suggesting a persistent effect of the neonatal diet. The sources of miRNAs in circulation need to be evaluated, as the piglets were fed the same solid diet leading up to PND 51 collections. In conclusion, the HM diet appears to have an immediate and persistent effect on the miRNA profile and likely regulates the pathways that impact the immune system and pose benefits to breastfed infants.
Collapse
Affiliation(s)
- Laura E Carr
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Ahmed A Elolimy
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Charity L Washam
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| | | | - Stewart L MacLeod
- Arkansas Children's Research Institute, Little Rock, AR, United States
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR, United States.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
180
|
Zeng B, Chen T, Luo J, Xie M, Wei L, Xi Q, Sun J, Zhang Y. Exploration of Long Non-coding RNAs and Circular RNAs in Porcine Milk Exosomes. Front Genet 2020; 11:652. [PMID: 32714373 PMCID: PMC7343709 DOI: 10.3389/fgene.2020.00652] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022] Open
Abstract
RNA in milk exosomes can be absorbed in the mammalian intestinal tract and function in gene expression regulations. Our previous work demonstrated that porcine milk exosomes (PME) contain large amounts of miRNAs and mRNAs. Increasing evidence suggests that long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are of particular interest, given their key role in diverse biological processes of animals. However, the expression profiles and the potential functions of lncRNAs and circRNAs in PME are still unknown. In the present study, we isolated PME by ultracentrifugation and performed a comprehensive analysis of lncRNA and circRNA in PME by using RNA sequencing. As a result, 2,466 novel lncRNAs, 809 annotated lncRNAs, and 61 circRNAs were identified in PME. The lncRNAs shared similar characteristics with other mammals in terms of length, exon number, and open reading frames. However, lncRNAs showed a higher level compared with mRNAs. Eight lncRNAs and five circRNAs in PME were selected for PCR identification. A functional enrichment analysis on the target genes of lncRNAs indicated that these genes were involved in cellular macromolecule metabolic, RNA metabolic, and immune processes. The circRNAs host genes were enriched in nucleic acid binding and adherence junction. We also evaluated the potential interaction targets between miRNAs and PME lncRNAs or circRNAs, and the results showed that the PME lncRNAs and the circRNAs have a high density of miRNA target sites. The top 20 highly expressed lncRNAs were found to interact with the proliferation-related miRNAs, and the circRNAs potentially targeted many miRNAs that are associated with the intestinal barrier. This study is the first to provide a resource for lncRNA and circRNA research of porcine milk. Moreover, the potential interaction between lncRNA/circRNA and miRNA is revealed. The present study expands our knowledge of non-coding RNAs in milk, and additional research is necessary to confirm their exactly physiological functions.
Collapse
Affiliation(s)
- Bin Zeng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Meiying Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Limin Wei
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
181
|
|
182
|
Wang Y, Zhang Y, Cai G, Li Q. Exosomes as Actively Targeted Nanocarriers for Cancer Therapy. Int J Nanomedicine 2020; 15:4257-4273. [PMID: 32606676 PMCID: PMC7306454 DOI: 10.2147/ijn.s239548] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
In recent years, it has been found that exosomes can be used as nanocarriers, which can be used in the treatment of tumors by carrying contents. The exosomes are derived from the secretion of the organism's own cells and are characterized by a phospholipid bilayer structure and a small particle size. These characteristics guarantee that the exosomes can carry a wide range of tumor drugs, deliver the drug to the cancer, and reduce or eliminate the tumor drug band. The toxic side effects were significantly eliminated; meanwhile, the therapeutic effects of the drug on the tumor were remarkably improved. This paper reviewed the strategies and drugs presented by different scholars for the treatment of tumors based on the drugs carried by exosomes.
Collapse
Affiliation(s)
- Yan Wang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Yingru Zhang
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Gang Cai
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| | - Qi Li
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai201203, People’s Republic of China
| |
Collapse
|
183
|
Matic S, D’Souza DH, Wu T, Pangloli P, Dia VP. Bovine Milk Exosomes Affect Proliferation and Protect Macrophages against Cisplatin-Induced Cytotoxicity. Immunol Invest 2020; 49:711-725. [DOI: 10.1080/08820139.2020.1769647] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Svjetlana Matic
- Department of Food Science, University of Tennessee, Knoxville, Tennessee, USA
| | - Doris H. D’Souza
- Department of Food Science, University of Tennessee, Knoxville, Tennessee, USA
| | - Tao Wu
- Department of Food Science, University of Tennessee, Knoxville, Tennessee, USA
| | - Philipus Pangloli
- Department of Food Science, University of Tennessee, Knoxville, Tennessee, USA
| | - Vermont P. Dia
- Department of Food Science, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
184
|
Stremmel W, Weiskirchen R, Melnik BC. Milk Exosomes Prevent Intestinal Inflammation in a Genetic Mouse Model of Ulcerative Colitis: A Pilot Experiment. Inflamm Intest Dis 2020; 5:117-123. [PMID: 32999884 DOI: 10.1159/000507626] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/30/2020] [Indexed: 12/17/2022] Open
Abstract
Background Milk is rich in nutrients and anabolic mediators rendering it essential for postnatal growth and metabolic programming. However, in adults, excessive consumption of milk is controversial as civilization disorders such as diabetes or prostate cancer may be promoted. A cytoprotective effect of milk could be utilized in inflammatory conditions, that is, chronic colitis. Objective To evaluate the effect of bovine milk exosomes on intestinal inflammation in a genetic mouse model of ulcerative colitis. Methods Intestinal-specific kindlin 2 knockout (KO) mice were exposed for 4 days to tamoxifen for induction of an ulcerative colitis phenotype. At the same time 4 other kindlin 2 KO mice were exposed to 33 μg/g cow milk derived exosomes in PBS by oral gavage. Both groups were compared to untreated wild-type controls. Results Milk exosomes prevented the appearance of a severe ulcerative phenotype. The macroscopic colitis score dropped from a mean of 3.33 in untreated mice to 0.75 index points (p < 0.01) in exosome-treated mice, which included significant improvement of the subscores of stool improvement and colon weight and length. Treated mice featured a noninflamed appearance of the intestinal mucosa. Key Message Milk exosomes have cytoprotective/anti-inflammatory activity in a genetic mouse model of ulcerative colitis. The mechanisms behind this need to be elucidated. This pilot study needs verification before a therapeutic strategy is developed.
Collapse
Affiliation(s)
- Wolfgang Stremmel
- Department of Gastroenterology, Medical Center Baden-Baden, Baden-Baden, Germany
| | - Ralf Weiskirchen
- Experimental Gene Therapy and Clinical Chemistry, Institute of Molecular Pathobiochemistry, RWTH University Hospital Aachen, Aachen, Germany
| | - Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
185
|
Sanwlani R, Fonseka P, Chitti SV, Mathivanan S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes 2020; 8:11. [PMID: 32414045 PMCID: PMC7356197 DOI: 10.3390/proteomes8020011] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Milk is considered as more than a source of nutrition for infants and is a vector involved in the transfer of bioactive compounds and cells. Milk contains abundant quantities of extracellular vesicles (EVs) that may originate from multiple cellular sources. These nanosized vesicles have been well characterized and are known to carry a diverse cargo of proteins, nucleic acids, lipids and other biomolecules. Milk-derived EVs have been demonstrated to survive harsh and degrading conditions in gut, taken up by various cell types, cross biological barriers and reach peripheral tissues. The cargo carried by these dietary EVs has been suggested to have a role in cell growth, development, immune modulation and regulation. Hence, there is considerable interest in understanding the role of milk-derived EVs in mediating inter-organismal and cross-species communication. Furthermore, various attributes such as it being a natural source, as well as its abundance, scalability, economic viability and lack of unwarranted immunologic reactions, has generated significant interest in deploying milk-derived EVs for clinical applications such as drug delivery and disease therapy. In this review, the role of milk-derived EVs in inter-organismal, cross-species communication and in drug delivery is discussed.
Collapse
Affiliation(s)
| | | | | | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia; (R.S.); (P.F.); (S.V.C.)
| |
Collapse
|
186
|
Lyons KE, Ryan CA, Dempsey EM, Ross RP, Stanton C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020; 12:E1039. [PMID: 32283875 PMCID: PMC7231147 DOI: 10.3390/nu12041039] [Citation(s) in RCA: 266] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/27/2022] Open
Abstract
Human breast milk is considered the optimum feeding regime for newborn infants due to its ability to provide complete nutrition and many bioactive health factors. Breast feeding is associated with improved infant health and immune development, less incidences of gastrointestinal disease and lower mortality rates than formula fed infants. As well as providing fundamental nutrients to the growing infant, breast milk is a source of commensal bacteria which further enhance infant health by preventing pathogen adhesion and promoting gut colonisation of beneficial microbes. While breast milk was initially considered a sterile fluid and microbes isolated were considered contaminants, it is now widely accepted that breast milk is home to its own unique microbiome. The origins of bacteria in breast milk have been subject to much debate, however, the possibility of an entero-mammary pathway allowing for transfer of microbes from maternal gut to the mammary gland is one potential pathway. Human milk derived strains can be regarded as potential probiotics; therefore, many studies have focused on isolating strains from milk for subsequent use in infant health and nutrition markets. This review aims to discuss mammary gland development in preparation for lactation as well as explore the microbial composition and origins of the human milk microbiota with a focus on probiotic development.
Collapse
Affiliation(s)
- Katríona E. Lyons
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
| | - Eugene M. Dempsey
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
- INFANT Research Centre, University College Cork, Cork T12 DFK4, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
187
|
Stephen BJ, Pareek N, Saeed M, Kausar MA, Rahman S, Datta M. Xeno-miRNA in Maternal-Infant Immune Crosstalk: An Aid to Disease Alleviation. Front Immunol 2020; 11:404. [PMID: 32269563 PMCID: PMC7109445 DOI: 10.3389/fimmu.2020.00404] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk is a complex liquid that contains multifaceted compounds which provide nutrition to infants and helps to develop their immune system. The presence of secretory immunoglobulins (IgA), leucocytes, lysozyme, lactoferrin, etc., in breast milk and their role in imparting passive immunity to infants as well as modulating development of an infant's immune system is well-established. Breast milk miRNAs (microRNAs) have been found to be differentially expressed in diverse tissues and biological processes during various molecular functions. Lactation is reported to assist mothers and their offspring to adapt to an ever-changing food supply. It has been observed that certain subtypes of miRNAs exist that are codified by non-human genomes but are still present in circulation. They have been termed as xeno-miRNA (XenomiRs). XenomiRs in humans have been found from various exogenous sources. Route of entry in human systems have been mainly dietary. The possibility of miRNAs taken up into mammalian circulation through diet, and thereby effecting gene expression, is a distinct possibility. This mechanism suggests an interesting possibility that dietary foods may modulate the immune strength of infants via highly specific post-transcriptional regulatory information present in mother's milk. This serves as a major breakthrough in understanding the fundamentals of nutrition and cross-organism communication. In this review, we elaborate and understand the complex crosstalk of XenomiRs present in mother's milk and their plausible role in modulating the infant immune system against infectious and inflammatory diseases.
Collapse
Affiliation(s)
| | - Nidhi Pareek
- Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicines, University of Ha'il, Ha'il, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, Munshi Singh College, Babasaheb Bhimrao Ambedkar Bihar University, Muzaffarpur, India
| | - Manali Datta
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
188
|
Galley JD, Besner GE. The Therapeutic Potential of Breast Milk-Derived Extracellular Vesicles. Nutrients 2020; 12:nu12030745. [PMID: 32168961 PMCID: PMC7146576 DOI: 10.3390/nu12030745] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/21/2022] Open
Abstract
In the past few decades, interest in the therapeutic benefits of exosomes and extracellular vesicles (EVs) has grown exponentially. Exosomes/EVs are small particles which are produced and exocytosed by cells throughout the body. They are loaded with active regulatory and stimulatory molecules from the parent cell including miRNAs and enzymes, making them prime targets in therapeutics and diagnostics. Breast milk, known for years to have beneficial health effects, contains a population of EVs which may mediate its therapeutic effects. This review offers an update on the therapeutic potential of exosomes/EVs in disease, with a focus on EVs present in human breast milk and their remedial effect in the gastrointestinal disease necrotizing enterocolitis. Additionally, the relationship between EV miRNAs, health, and disease will be examined, along with the potential for EVs and their miRNAs to be engineered for targeted treatments.
Collapse
|
189
|
Carobolante G, Mantaj J, Ferrari E, Vllasaliu D. Cow Milk and Intestinal Epithelial Cell-derived Extracellular Vesicles as Systems for Enhancing Oral Drug Delivery. Pharmaceutics 2020; 12:E226. [PMID: 32143503 PMCID: PMC7150822 DOI: 10.3390/pharmaceutics12030226] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022] Open
Abstract
Ingestion is the preferred way for drug administration. However, many drugs have poor oral bioavailability, warranting the use of injections. Extracellular vesicles (EVs) from cow milk have shown potential utility in improving oral drug bioavailability. However, EVs produced by intestinal epithelial cells have not been investigated for this application. We compared the capacity of cow milk EVs and intestinal epithelial cell-derived counterparts to enhance oral drug bioavailability. EVs were isolated, fluorescently labelled, and loaded with curcumin (CUR) as a model poorly absorbable drug. These were then characterised before testing in an intestinal model (Caco-2). Epithelial cell-derived EVs showed notably higher cell uptake compared to cow milk EVs. Cell uptake was significantly higher in differentiated compared to undifferentiated cells for both types of EVs. While both milk- and cell-derived EVs improved the cell uptake and intestinal permeability of CUR (confirming oral drug bioavailability enhancement potential), epithelial cell EVs demonstrated a superior effect.
Collapse
Affiliation(s)
- Greta Carobolante
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9NH, UK; (G.C.); (J.M.)
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di PADOVA, Via 8 Febbraio, 2, 35122 Padova, Italy
| | - Julia Mantaj
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9NH, UK; (G.C.); (J.M.)
| | - Enrico Ferrari
- School of Life Sciences, University of Lincoln, Lincoln LN6 7TS, UK;
| | - Driton Vllasaliu
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE1 9NH, UK; (G.C.); (J.M.)
| |
Collapse
|
190
|
Xie MY, Chen T, Xi QY, Hou LJ, Luo JY, Zeng B, Li M, Sun JJ, Zhang YL. Porcine milk exosome miRNAs protect intestinal epithelial cells against deoxynivalenol-induced damage. Biochem Pharmacol 2020; 175:113898. [PMID: 32145262 DOI: 10.1016/j.bcp.2020.113898] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/02/2020] [Indexed: 12/18/2022]
Abstract
Porcine milk exosomes play an important role in mother-infant communication. Deoxynivalenol (DON) is a toxin which causes serious damage to the animal intestinal mucosa. Our previous study showed porcine milk exosomes facilitate mice intestine development, but the effects of these exosomes to antagonize DON toxicity is unclear. Our in vivo results showed that milk exosomes attenuated DON-induced damage on the mouse body weight and intestinal epithelium growth. In addition, these exosomes could reverse DON-induced inhibition on cell proliferation and tight junction proteins (TJs) formation and reduce DON-induced cell apoptosis. In vitro, exosomes up-regulated the expression of miR-181a, miR-30c, miR-365-5p and miR-769-3p in IPEC-J2 cells and then down-regulated the expression of their targeting genes in p53 pathway, ultimately attenuating DON-induced damage by promoting cell proliferation and TJs and by inhibiting cell apoptosis. In conclusion, porcine milk exosomes could protect the intestine against DON damage, and these protections may take place through the miRNAs in exosomes. These results indicated that the addition of miRNA-enriched exosomes to feed or food could be used as a novel preventative measure for necrotizing enterocolitis.
Collapse
Affiliation(s)
- Mei-Ying Xie
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Collaborative Innovation Center of Plant Pest Management and Bioenvironmental Health Application Technology, Guangdong Eco-Engineering Polytechnic, 297 Guangshan First Road, Tianhe District, Guangzhou, Guangdong 510520, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Ting Chen
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Engineering&Research Center for Woody Fodder Plants, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Qian-Yun Xi
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Engineering&Research Center for Woody Fodder Plants, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Lian-Jie Hou
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Jun-Yi Luo
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Bin Zeng
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Meng Li
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China
| | - Jia-Jie Sun
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Engineering&Research Center for Woody Fodder Plants, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China.
| | - Yong-Liang Zhang
- College of Animal Science, South China Agricultural University, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Province Key Laboratory of Animal Nutritional Regulation, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; National Engineering Research Center for Breeding Swine Industry, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China; Guangdong Engineering&Research Center for Woody Fodder Plants, 483 Wushan Road, Tianhe District, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
191
|
Kirchner B, Buschmann D, Paul V, Pfaffl MW. Postprandial transfer of colostral extracellular vesicles and their protein and miRNA cargo in neonatal calves. PLoS One 2020; 15:e0229606. [PMID: 32109241 PMCID: PMC7048281 DOI: 10.1371/journal.pone.0229606] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) such as exosomes are key regulators of intercellular communication that can be found in almost all bio fluids. Although studies in the last decade have made great headway in discerning the role of EVs in many physiological and pathophysiological processes, the bioavailability and impact of dietary EVs and their cargo still remain to be elucidated. Due to its widespread consumption and high content of EV-associated microRNAs and proteins, a major focus in this field has been set on EVs in bovine milk and colostrum. Despite promising in vitro studies in recent years that show high resiliency of milk EVs to degradation and uptake of milk EV cargo in a variety of intestinal and blood cell types, in vivo experiments continue to be inconclusive and sometimes outright contradictive. To resolve this discrepancy, we assessed the potential postprandial transfer of colostral EVs to the circulation of newborn calves by analysing colostrum-specific protein and miRNAs, including specific isoforms (isomiRs) in cells, EV isolations and unfractionated samples from blood and colostrum. Our findings reveal distinct populations of EVs in colostrum and blood from cows that can be clearly separated by density, particle concentration and protein content (BTN1A1, MFGE8). Postprandial blood samples of calves show a time-dependent increase in EVs that share morphological and protein characteristics of colostral EVs. Analysis of miRNA expression profiles by Next-Generation Sequencing gave a different picture however. Although significant postprandial expression changes could only be detected for calf EV samples, expression profiles show very limited overlap with highly expressed miRNAs in colostral EVs or colostrum in general. Taken together our results indicate a selective uptake of membrane-associated protein cargo but not luminal miRNAs from colostral EVs into the circulation of neonatal calves.
Collapse
Affiliation(s)
- Benedikt Kirchner
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
- * E-mail:
| | - Dominik Buschmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
- Institute of Human Genetics, University Hospital, LMU Munich, Munich, Germany
| | - Vijay Paul
- National Research Centre on Yak, ICAR, Dirang, India
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| |
Collapse
|
192
|
Extracellular Vesicles with Possible Roles in Gut Intestinal Tract Homeostasis and IBD. Mediators Inflamm 2020; 2020:1945832. [PMID: 32410847 PMCID: PMC7201673 DOI: 10.1155/2020/1945832] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/17/2019] [Accepted: 12/24/2019] [Indexed: 12/17/2022] Open
Abstract
The intestinal tract consists of various types of cells, such as epithelial cells, Paneth cells, macrophages, and lymphocytes, which constitute the intestinal immune system and play a significant role in maintaining intestinal homeostasis by producing antimicrobial materials and controlling the host-commensal balance. Various studies have found that the dysfunction of intestinal homeostasis contributes to the pathogenesis of inflammatory bowel disease (IBD). As a novel mediator, extracellular vesicles (EVs) have been recognized as effective communicators, not only between cells but also between cells and the organism. In recent years, EVs have been regarded as vital characters for dysregulated homeostasis and IBD in either the etiology or the pathology of intestinal inflammation. Here, we review recent studies on EVs associated with intestinal homeostasis and IBD and discuss their source, cargo, and origin, as well as their therapeutic effects on IBD, which mainly include artificial nanoparticles and EVs derived from microorganisms.
Collapse
|
193
|
Pisano C, Galley J, Elbahrawy M, Wang Y, Farrell A, Brigstock D, Besner GE. Human Breast Milk-Derived Extracellular Vesicles in the Protection Against Experimental Necrotizing Enterocolitis. J Pediatr Surg 2020; 55:54-58. [PMID: 31685268 PMCID: PMC6989376 DOI: 10.1016/j.jpedsurg.2019.09.052] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 09/29/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) is a leading cause of death in premature infants. Breast feeding decreases the incidence of NEC but, even with aggressive promotion of nursing in Neonatal Intensive Care Units, morbidity and mortality remain high. Previous studies from our laboratory have demonstrated that extracellular vesicles (EVs) purified from mouse and rat stem cells can protect the intestines from NEC. The aim of this study was to determine whether human breast milk (BM)-derived EVs could prevent NEC. METHODS EVs were purified from human donor breast milk. NEC was induced in premature rat pups by exposure to asphyxia/hypothermia/hypercaloric feeds. Pups were randomized to: (1) breast fed, no injury, (2) NEC, (3) NEC + BM-derived EVs once intraperitoneally (IP), (4) NEC + BM-derived EVs enterally (PO) with each feed. Intestinal tracts were examined for histologic damage. Additionally, the effect of BM-derived EVs on intestinal epithelial cells (IEC) subjected to hypoxia/reoxygenation injury in vitro was examined. RESULTS NEC incidence was 0% in breast-fed pups and 62% in pups subjected to NEC. IP administration of BM-derived EVs decreased NEC incidence to 29% and enteral administration further decreased NEC incidence to 11.9%. (p < 0.05). BM-derived EVs significantly increased cell proliferation and decreased apoptosis in IEC in vitro. CONCLUSION Breast milk-derived EVs delivered either IP or enterally significantly decrease the incidence and severity of experimental NEC, protect IEC from injury in vitro, and may represent an innovative therapeutic option for NEC in the future. TYPE OF STUDY Basic science study. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Courtney Pisano
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - Jeffrey Galley
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - Mostafa Elbahrawy
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - Yijie Wang
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - Aidan Farrell
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - David Brigstock
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH
| | - Gail E Besner
- Department of Pediatric Surgery, Center for Perinatal Research Nationwide Children's Hospital, Columbus, OH.
| |
Collapse
|
194
|
Kakimoto Y, Matsushima Y, Tanaka M, Hayashi H, Wang T, Yokoyama K, Ochiai E, Osawa M. MicroRNA profiling of gastric content from breast-fed and formula-fed infants to estimate last feeding: a pilot study. Int J Legal Med 2019; 134:903-909. [PMID: 31832755 DOI: 10.1007/s00414-019-02226-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/04/2019] [Indexed: 02/06/2023]
Abstract
Recently, we were consulted about a challenging case, where an infant died by poisoning and the drug-dependent mother insisted that she unintentionally gave the toxic drug through breast milk. Accordingly, we investigated the utility of immunoblotting and microRNA (miRNA) profiling of the infant's gastric content (GC) to differentiate between breast-feeding and formula-feeding. As a pilot study, we sampled the GC from breast-fed (GCB) and formula-fed (GCF) infants, as well as gastric juice (GJ) from fasted adults at autopsy. Breast milk (BM) samples were collected from volunteers within 1 year post-delivery. By immunoblotting, lactoferrin and gross cystic disease fluid protein (GCDEP) were clearly detected in BM, but could not be detected in GCB. Similarly, β-lactoglobulin was detected in formula milk, but could not be detected in GCF. Meanwhile, miRNA sequencing revealed that the miRNA expression profile of GCB was closer to BM than GCF and GJ. Especially, miR-151a and miR-186 were more abundant in BM and GCB than in GCF and GJ. Our study is the first to elucidate the human GJ miRNA profile and demonstrate the possibility that miR-151a and miR-186 in GC may be the biomarker of breast-feeding.
Collapse
Affiliation(s)
- Yu Kakimoto
- Department of Forensic Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan.
| | - Yutaka Matsushima
- Department of Forensic Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Masayuki Tanaka
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Hideki Hayashi
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Ting Wang
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Keiko Yokoyama
- Support Center for Medical Research and Education, Tokai University, Kanagawa, Japan
| | - Eriko Ochiai
- Department of Forensic Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| | - Motoki Osawa
- Department of Forensic Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1193, Japan
| |
Collapse
|
195
|
Li J, Lei L, Ye F, Zhou Y, Chang H, Zhao G. Nutritive implications of dietary microRNAs: facts, controversies, and perspectives. Food Funct 2019; 10:3044-3056. [PMID: 31066412 DOI: 10.1039/c9fo00216b] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
As a group of non-coding RNA molecules, microRNAs have recently become more well-known due to their pivotal role in gene regulation. A large number of endogenous microRNAs naturally occur in the human body, and some of them act as regulatory targets of diet and its components. The wide presence of microRNAs in various food materials has inspired food scientists and nutritionists to explore their nutritive and bioactive significance. This article comprehensively reports updated insights into the accessibility, stability, absorbability, and bioactivity of dietary microRNAs by combining the current knowledge into figures and tables for reader's convenience. As one frontier in food science and nutrition, the research platform on dietary microRNAs is imperfect and even defective as indicated by the inconsistent and even contradictory results concluded by different investigations. The pros and cons as well as the limitations of current investigations have been critically discussed with attention chiefly paid to experimental designs and protocols. Moreover, future research directions have been recommended. Thus, this paper may not only provide a quick glance at the state-of-the-art of dietary microRNAs but also guide further research to clarify the present controversies and make the results more credible and persuasive.
Collapse
Affiliation(s)
- Jianting Li
- College of Food Science, Southwest University, Chongqing 400715, People's Republic of China.
| | | | | | | | | | | |
Collapse
|
196
|
Gao R, Zhang R, Qian T, Peng X, He W, Zheng S, Cao Y, Pierro A, Shen C. A comparison of exosomes derived from different periods breast milk on protecting against intestinal organoid injury. Pediatr Surg Int 2019; 35:1363-1368. [PMID: 31576466 DOI: 10.1007/s00383-019-04562-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2019] [Indexed: 02/05/2023]
Abstract
AIM OF THE STUDY Human breast milk reduces the risk and severity of necrotizing enterocolitis (NEC). Exosomes are extracellular vesicles (EVs) found in high concentrations in milk, and they mediate intercellular communication and immune responses. The aim of this study is to compare the protective effects of exosomes that are derived from different time periods of breast milk production against intestinal injury using an ex vivo intestinal organoid model. METHODS Colostrum, transitional and mature breast milk samples from healthy lactating mothers were collected. Exosomes were isolated using serial ultracentrifugation and filtration. Exosomes' presence was confirmed using transmission electron microscopy (TEM) and western blot. To form the intestinal organoids, terminal ileum was harvested from neonatal mice pups at postnatal day 9, crypts were isolated and organoids were cultured in matrigel. Organoids were either cultured with exposure to lipopolysaccharide (LPS), or in treatment groups where both LPS and exosomes were added in the culturing medium. Inflammatory markers and organoids viability were evaluated. MAIN RESULTS Human milk-derived exosomes were successfully isolated and characterized. LPS administration reduced the size of intestinal organoids, induced inflammation through increasing TNFα and TLR4 expression, and stimulated intestinal regeneration. Colostrum, transitional and mature human milk-derived exosome treatment all prevented inflammatory injury, while exosomes derived from colostrum were most effective at reducing inflammatory cytokine. CONCLUSIONS Human breast milk-derived exosomes were able to protect intestine organoids against epithelial injury induced by LPS. Colostrum exosomes offer the best protective effect among the breast-milk derived exosomes. Human milk exosomes can be protective against the development of intestinal injury such as that seen in NEC.
Collapse
Affiliation(s)
- Runnan Gao
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan RD, Shanghai, 201102, China
| | - Rong Zhang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Tian Qian
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Xueni Peng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan RD, Shanghai, 201102, China
| | - Weijing He
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan RD, Shanghai, 201102, China
| | - Shan Zheng
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan RD, Shanghai, 201102, China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Agostino Pierro
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, ON, Canada
| | - Chun Shen
- Department of Pediatric Surgery, Children's Hospital of Fudan University, 399 Wanyuan RD, Shanghai, 201102, China.
| |
Collapse
|
197
|
Wu D, Kittana H, Shu J, Kachman SD, Cui J, Ramer-Tait AE, Zempleni J. Dietary Depletion of Milk Exosomes and Their MicroRNA Cargos Elicits a Depletion of miR-200a-3p and Elevated Intestinal Inflammation and Chemokine (C-X-C Motif) Ligand 9 Expression in Mdr1a-/- Mice. Curr Dev Nutr 2019; 3:nzz122. [PMID: 32154493 PMCID: PMC7053579 DOI: 10.1093/cdn/nzz122] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Exosomes transfer regulatory microRNAs (miRs) from donor cells to recipient cells. Exosomes and miRs originate from both endogenous synthesis and dietary sources such as milk. miR-200a-3p is a negative regulator of the proinflammatory chemokine (C-X-C motif) ligand 9 (CXCL9). Male Mdr1a-/- mice spontaneously develop clinical signs of inflammatory bowel disease (IBD). OBJECTIVES We assessed whether dietary depletion of exosomes and miRs alters the severity of IBD in Mdr1a-/- mice owing to aberrant regulation of proinflammatory cytokines. METHODS Starting at 5 wk of age, 16 male Mdr1a-/- mice were fed either milk exosome- and RNA-sufficient (ERS) or milk exosome- and RNA-depleted (ERD) diets. The ERD diet is characterized by a near-complete depletion of miRs and a 60% loss of exosome bioavailability compared with ERS. Mice were killed when their weight loss exceeded 15% of peak body weight. Severity of IBD was assessed by histopathological evaluation of cecum. Serum cytokine and chemokine concentrations and mRNA and miR tissue expression were analyzed by multiplex ELISAs, RNA-sequencing analysis, and qRT-PCR, respectively. RESULTS Stromal collapse, gland hyperplasia, and additive microscopic disease scores were (mean ± SD) 56.7% ± 23.3%, 23.5% ± 11.8%, and 29.6% ± 8.2% lower, respectively, in ceca of ERS mice than of ERD mice (P < 0.05). The serum concentration of CXCL9 was 35.0% ± 31.0% lower in ERS mice than in ERD mice (P < 0.05). Eighty-seven mRNAs were differentially expressed in the ceca from ERS and ERD mice; 16 of these mRNAs are implicated in immune function. The concentrations of 4 and 1 out of 5 miRs assessed (including miR-200a-3p) were ≤63% lower in livers and ceca, respectively, from ERD mice than from ERS mice. CONCLUSIONS Milk exosome and miR depletion exacerbates cecal inflammation in Mdr1a-/- mice.
Collapse
Affiliation(s)
- Di Wu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Hatem Kittana
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jiang Shu
- Department of Computer Science and Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Stephen D Kachman
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Juan Cui
- Department of Computer Science and Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
198
|
The Beneficial Effect of Farm Milk Consumption on Asthma, Allergies, and Infections: From Meta-Analysis of Evidence to Clinical Trial. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 8:878-889.e3. [PMID: 31770653 DOI: 10.1016/j.jaip.2019.11.017] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/23/2022]
Abstract
The low prevalence of asthma and allergies in farm children has partially been ascribed to the consumption of raw cow's milk. A literature search identified 12 publications on 8 pertinent studies. A meta-analysis corroborated the protective effect of raw milk consumption early in life (<1 to 5 years, according to study) on asthma (odds ratio [OR], 0.58; 95% CI, 0.49-0.69), current wheeze (OR, 0.66; 95% CI, 0.55-0.78), hay fever or allergic rhinitis (OR, 0.68; 95% CI, 0.57-0.82), and atopic sensitization (OR, 0.76; 95% CI, 0.62-0.95). The effect particularly on asthma was observed not only in children raised on farms (OR, 0.62; 95% CI, 0.58-0.82) but also in children living in rural areas but not on a farm (OR, 0.60; 95% CI, 0.48-0.74). This demonstrates that the effect of farm milk consumption is independent of other farm exposures and that children not living on a farm can theoretically profit from this effect. Because of the minimal but real risk of life-threatening infections, however, consumption of raw milk and products thereof is strongly discouraged. Raw farm milk and industrially processed milk differ in many instances including removal of cellular components, manipulation of the fat fraction, and various degrees of heating. Preliminary evidence attributes the effect to heat-labile molecules and components residing in the fat fraction. The Milk Against Respiratory Tract Infections and Asthma (MARTHA) trial is currently testing the protective effect of microbiologically safe, minimally processed cow's milk against standard ultra-heat-treated milk in children from 6 months to 3 years with the primary outcome of an asthma diagnosis until age 5 years. If successful, this approach might provide a simple but effective prevention strategy.
Collapse
|
199
|
Mirza AH, Kaur S, Nielsen LB, Størling J, Yarani R, Roursgaard M, Mathiesen ER, Damm P, Svare J, Mortensen HB, Pociot F. Breast Milk-Derived Extracellular Vesicles Enriched in Exosomes From Mothers With Type 1 Diabetes Contain Aberrant Levels of microRNAs. Front Immunol 2019; 10:2543. [PMID: 31708933 PMCID: PMC6823203 DOI: 10.3389/fimmu.2019.02543] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/14/2019] [Indexed: 12/12/2022] Open
Abstract
The breast milk plays a crucial role in shaping the initial intestinal microbiota and mucosal immunity of the infant. Interestingly, breastfeeding has proven to be protective against the early onset of immune-mediated diseases including type 1 diabetes. Studies have shown that exosomes from human breast milk are enriched in immune-modulating miRNAs suggesting that exosomal miRNAs (exomiRs) transferred to the infant could play a critical role in the development of the infant's immune system. We extracted exomiRs from breast milk of 52 lactating mothers (26 mothers with type 1 diabetes and 26 healthy mothers), to identify any differences in the exomiR content between the two groups. Small RNA-sequencing was performed to identify known and novel miRNAs in both groups. A total of 631 exomiRs were detected by small RNA sequencing including immune-related miRNAs such as hsa-let-7c, hsa-miR-21, hsa-miR-34a, hsa-miR-146b, and hsa-miR-200b. In addition, ~200 novel miRNAs were identified in both type 1 diabetes and control samples. Among the known miRNAs, nine exomiR's were found differentially expressed in mothers with type 1 diabetes compared to healthy mothers. The highly up-regulated miRNAs, hsa-miR-4497, and hsa-miR-3178, increased lipopolysaccharide-induced expression and secretion of tumor necrosis factor α (TNFα) in human monocytes. The up-regulated miRNA target genes were significantly enriched for longevity-regulating pathways and FoxO signaling. Our findings suggest a role of breast milk-derived exomiRs in modulating the infant's immune system.
Collapse
Affiliation(s)
- Aashiq H Mirza
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Simranjeet Kaur
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lotte B Nielsen
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Joachim Størling
- Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Reza Yarani
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Martin Roursgaard
- Faculty of Health Sciences, Institute of Public Health, CSS, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth R Mathiesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Damm
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Pregnant Women With Diabetes, Rigshospitalet, Copenhagen, Denmark.,Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Jens Svare
- Department of Obstetrics, Herlev Hospital, Herlev, Denmark
| | - Henrik B Mortensen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark
| | - Flemming Pociot
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pediatrics E, Copenhagen Diabetes Research Center (CPH-DIRECT), Herlev and Gentofte Hospital, Herlev, Denmark.,Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
200
|
Benmoussa A, Diallo I, Salem M, Michel S, Gilbert C, Sévigny J, Provost P. Concentrates of two subsets of extracellular vesicles from cow's milk modulate symptoms and inflammation in experimental colitis. Sci Rep 2019; 9:14661. [PMID: 31601878 PMCID: PMC6787204 DOI: 10.1038/s41598-019-51092-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
Extracellular vesicles (EVs) are involved in cell-to-cell communication and modulation of numerous physiological and pathological processes. EVs are found in large quantities in milk and contain several inflammation- and immunity-modulating proteins and microRNAs, through which they exert beneficial effects in several inflammatory disease models. Here, we investigated the effects of two EV subsets, concentrated from commercial cow's milk, on a murine model of colitis induced with dextran sodium sulfate (DSS). P35K EVs, isolated by ultracentrifugation at 35,000 g, and P100K EVs, isolated at 100,000 g, were previously characterized and administered by gavage to healthy and DSS-treated mice. P35K EVs and, to a lesser extent, P100K EVs improved several outcomes associated to DSS-induced colitis, modulated the gut microbiota, restored intestinal impermeability and replenished mucin secretion. Also, P35K EVs modulated innate immunity, while P100K EVs decreased inflammation through the downregulation of colitis-associated microRNAs, especially miR-125b, associated with a higher expression of the NFκB inhibitor TNFAIP3 (A20). These results suggest that different milk EV subsets may improve colitis outcomes through different, and possibly complementary, mechanisms. Further unveiling of these mechanisms might offer new opportunities for improving the life of patients with colitis and be of importance for milk processing, infant milk formulation and general public health.
Collapse
Affiliation(s)
- Abderrahim Benmoussa
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Idrissa Diallo
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Mabrouka Salem
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Sara Michel
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Caroline Gilbert
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Jean Sévigny
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada
| | - Patrick Provost
- CHUQ Research Center/CHUL Pavilion, 2705 Blvd Laurier, Quebec, QC, G1V 4G2, Canada.
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Université Laval, Quebec, QC, G1V 0A6, Canada.
| |
Collapse
|