1
|
Saleem A, Saleem Bhat S, A. Omonijo F, A Ganai N, M. Ibeagha-Awemu E, Mudasir Ahmad S. Immunotherapy in mastitis: state of knowledge, research gaps and way forward. Vet Q 2024; 44:1-23. [PMID: 38973225 PMCID: PMC11232650 DOI: 10.1080/01652176.2024.2363626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/27/2024] [Indexed: 07/09/2024] Open
Abstract
Mastitis is an inflammatory condition that affects dairy cow's mammary glands. Traditional treatment approaches with antibiotics are increasingly leading to challenging scenarios such as antimicrobial resistance. In order to mitigate the unwanted side effects of antibiotics, alternative strategies such as those that harness the host immune system response, also known as immunotherapy, have been implemented. Immunotherapy approaches to treat bovine mastitis aims to enhance the cow's immune response against pathogens by promoting pathogen clearance, and facilitating tissue repair. Various studies have demonstrated the potential of immunotherapy for reducing the incidence, duration and severity of mastitis. Nevertheless, majority of reported therapies are lacking in specificity hampering their broad application to treat mastitis. Meanwhile, advancements in mastitis immunotherapy hold great promise for the dairy industry, with potential to provide effective and sustainable alternatives to traditional antibiotic-based approaches. This review synthesizes immunotherapy strategies, their current understanding and potential future perspectives. The future perspectives should focus on the development of precision immunotherapies tailored to address individual pathogens/group of pathogens, development of combination therapies to address antimicrobial resistance, and the integration of nano- and omics technologies. By addressing research gaps, the field of mastitis immunotherapy can make significant strides in the control, treatment and prevention of mastitis, ultimately benefiting both animal and human health/welfare, and environment health.
Collapse
Affiliation(s)
- Afnan Saleem
- Division of Animal Biotechnology, SKUAST-K, Srinagar, India
| | | | - Faith A. Omonijo
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | | - Eveline M. Ibeagha-Awemu
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, Canada
| | | |
Collapse
|
2
|
Çelik E, Cemali Ö, Şahin TÖ, Deveci G, Biçer NÇ, Hirfanoğlu İM, Ağagündüz D, Budán F. Human Breast Milk Exosomes: Affecting Factors, Their Possible Health Outcomes, and Future Directions in Dietetics. Nutrients 2024; 16:3519. [PMID: 39458514 PMCID: PMC11510026 DOI: 10.3390/nu16203519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Human breast milk is a complex biological fluid containing multifaceted biological compounds that boost immune and metabolic system development that support the short- and long-term health of newborns. Recent literature suggests that human breast milk is a substantial source of nutrients, bioactive molecules, and exosomes. Objectives: This review examines the factors influencing exosomes noted in human milk and the impacts of exosomes on infant health. Furthermore, it discusses potential future prospects for exosome research in dietetics. Methods: Through a narrative review of the existing literature, we focused on exosomes in breast milk, exosome components and their potential impact on exosome health. Results: Exosomes are single-membrane extracellular vesicles of endosomal origin, with an approximate radius of 20-200 nm. They are natural messengers that cells secrete to transport a wide range of diverse cargoes, including deoxyribonucleic acid, ribonucleic acid, proteins, and lipids between various cells. Some studies have reported that the components noted in exosomes in human breast milk could be transferred to the infant and cause epigenetic changes. Thus, it can affect gene expression and cellular event regulation in several tissues. Conclusions: In this manner, exosomes are associated with several pathways, including the immune system, oxidative stress, and cell cycle, and they can affect the short- and long-term health of infants. However, there is still much to learn about the functions, effectiveness, and certain impacts on the health of human breast milk exosomes.
Collapse
Affiliation(s)
- Elif Çelik
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Süleyman Demirel University, Isparta 32260, Türkiye;
| | - Özge Cemali
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Trakya University, Edirne 22030, Türkiye;
| | - Teslime Özge Şahin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Türkiye;
| | - Gülsüm Deveci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Çankırı Karatekin University, Çankırı 18100, Türkiye;
| | - Nihan Çakır Biçer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul 34752, Türkiye;
| | | | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Türkiye
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
3
|
Munir J, Sadri M, Zempleni J. Tsg101 knockout in the mammary gland leads to a decrease in small extracellular vesicles in milk from C57BL/6J dams and contributes to leakiness of the gut mucosa and reduced postnatal weight gain in suckling pups. J Nutr Biochem 2024; 135:109782. [PMID: 39424203 DOI: 10.1016/j.jnutbio.2024.109782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/21/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
Human milk contains 2.2 ± 1.5×1011 small extracellular vesicles (sEVs) per milliliter and human infants consume 1.7×1014 milk sEVs (sMEVs) daily in 800 mL milk. Infant formula contains trace amounts of sMEVs. To date, eight adverse effects of milk depletion and five beneficial effects of sMEV supplementation have been reported including studies in infants and neonate mice. Formula-fed infants do not realize the benefits of sMEVs. Most of the phenotyping studies reported to date have the limitation that sMEV depletion and supplementation were initiated after mice were weaned. Here, we used a genetics approach for assessing effects of sMEV depletion on the development of suckling mice. Newborn C57BL/6J pups were fostered to Tumor Susceptibility Gene 101 (Tsg101) mammary-specific knockout (KO) dams or C57BL/6J dams (controls) in synchronized pregnancies. Tsg101 KO was associated with an 80% decrease of sMEVs. Postnatal weight gain and gut health (histology, morphology, and barrier function) were assessed until weaning at age three weeks. We observed a significant decrease in weight gain, length of small intestine, villi height, crypt depth, and intestinal barrier function in male and female pups fostered to Tsg101 dams compared to pups fostered to control dams. The effect size varied between 11 and 32 percent. Maternal Tsg101 KO did not affect the dams' health, content of macronutrients and dry mass of milk and had no effect on the amount of milk consumed by pups. We conclude that sMEVs are important for growth and gut health in neonate mice.
Collapse
Affiliation(s)
- Javaria Munir
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Mahrou Sadri
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
4
|
Bui HTD, You G, Lee M, Mao W, So C, Byeon C, Hong S, Mok H, Yoo HS. Milk exosome-infused fibrous matrix for treatment of acute wound. J Control Release 2024; 376:79-93. [PMID: 39366455 DOI: 10.1016/j.jconrel.2024.09.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
To provide an advanced therapy for wound recovery, in this study, pasteurized bovine milk-derived exosomes (mEXO) are immobilized onto a polydopamine (PDA)-coated hyaluronic acid (HA)-based electrospun nanofibrous matrix (mEXO@PMAT) via a simple dip-coating method to formulate an mEXO-immobilized mesh as a wound-healing biomaterial. Purified mEXOs (∼82 nm) contain various anti-inflammatory, cell proliferation, and collagen synthesis-related microRNAs (miRNAs), including let-7b, miR-184, and miR-181a, which elicit elevated mRNA expression of keratin5, keratin14, and collagen1 in human keratinocytes (HaCaT) and fibroblasts (HDF). The mEXOs immobilized onto the PDA-coated meshes are gradually released from the meshes over 14 days without burst-out effect. After treatment with HaCaT and HDF, the degree of in vitro cell migration increases significantly in the mEXO@PMAT-treated HaCaT and HDF cells compared to the unmodified or PDA-coated meshes-treated cells. Additionally, the mEXO@PMAT provides significantly faster wound closure in vivo without notable toxicity. Thus, the sustained liberation of bioactive mEXO from the meshes can effectively enhance cell proliferation in vitro and accelerate wound closure in vivo, which could be harnessed mEXO@PMAT as a promising wound-healing biomaterial.
Collapse
Affiliation(s)
- Hoai-Thuong Duc Bui
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Gayeon You
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Miso Lee
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Wei Mao
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Chaewon So
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea
| | - Chorok Byeon
- Department of Physics and Chemistry, DGIST, Daegu, 42988, Republic of Korea
| | - Seonki Hong
- Department of Physics and Chemistry, DGIST, Daegu, 42988, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 143-701, Republic of Korea.
| | - Hyuk Sang Yoo
- Department of Medical Biomaterials Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute for Molecular Science and Fusion Technology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea; Kangwon Radiation Convergence Research Center, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
5
|
Butz H, Patócs A, Igaz P. Circulating non-coding RNA biomarkers of endocrine tumours. Nat Rev Endocrinol 2024; 20:600-614. [PMID: 38886617 DOI: 10.1038/s41574-024-01005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Circulating non-coding RNA (ncRNA) molecules are being investigated as biomarkers of malignancy, prognosis and follow-up in several neoplasms, including endocrine tumours of the pituitary, parathyroid, pancreas and adrenal glands. Most of these tumours are classified as neuroendocrine neoplasms (comprised of neuroendocrine tumours and neuroendocrine carcinomas) and include tumours of variable aggressivity. We consider them together here in this Review owing to similarities in their clinical presentation, pathomechanism and genetic background. No preoperative biomarkers of malignancy are available for several forms of these endocrine tumours. Moreover, biomarkers are also needed for the follow-up of tumour progression (especially in hormonally inactive tumours), prognosis and treatment efficacy monitoring. Circulating blood-borne ncRNAs show promising utility as biomarkers. These ncRNAs, including microRNAs, long non-coding RNAs and circular RNAs, are involved in several aspects of gene expression regulation, and their stability and tissue-specific expression could make them ideal biomarkers. However, no circulating ncRNA biomarkers have yet been introduced into routine clinical practice, which is mostly owing to methodological and standardization problems. In this Review, following a brief synopsis of these endocrine tumours and the biology of ncRNAs, the major research findings, pathomechanisms and methodological questions are discussed along with an outlook for future studies.
Collapse
Affiliation(s)
- Henriett Butz
- HUN-REN-SU Hereditary Tumours Research Group, Budapest, Hungary
- Department of Molecular Genetics and the National Tumour Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Patócs
- HUN-REN-SU Hereditary Tumours Research Group, Budapest, Hungary
- Department of Molecular Genetics and the National Tumour Biology Laboratory, National Institute of Oncology, Budapest, Hungary
- Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter Igaz
- Department of Endocrinology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
- Department of Internal Medicine and Oncology, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
6
|
Lu X, Ren K, Pan L, Liu X. Sheep ( Ovis aries) Milk Exosomal miRNAs Attenuate Dextran Sulfate Sodium-Induced Colitis in Mice via TLR4 and TRAF-1 Inhibition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:21030-21040. [PMID: 39283309 DOI: 10.1021/acs.jafc.4c05524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Mammalian milk exosomal miRNAs play an important role in maintaining intestinal immune homeostasis and protecting epithelial barrier function, but the specific miRNAs and whether miRNA-mediated mechanisms are responsible for these benefits remain a matter of investigation. This study isolated sheep milk-derived exosomes (sheep MDEs), identifying the enriched miRNAs in sheep MDEs, oar-miR-148a, and oar-let-7b as key components targeting TLR4 and TRAF1, which was validated by a dual-luciferase reporter assay. In dextran sulfate sodium-induced colitis mice, administration of sheep MDEs alleviated colitis symptoms, reduced colonic inflammation, and systemic oxidative stress, as well as significantly increased colonic oar-miR-148a and oar-let-7b while reducing toll-like receptor 4 (TLR4) and TNF-receptor-associated factor 1 (TRAF1) level. Further characterization in TNF-α-challenged Caco-2 cells showed that overexpression of these miRNAs suppressed the TLR4/TRAF1-IκBα-p65 pathway and reduced IL-6 and IL-12 production. These findings indicate that sheep MDEs exert gastrointestinal anti-inflammatory effects through the miRNA-mediated modulation of TLR4 and TRAF1, highlighting their potential in managing colitis.
Collapse
Affiliation(s)
- Xi Lu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710000, China
| | - Ke Ren
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710000, China
| | - Lei Pan
- Tangdu Hospital, Air Force Military Medical University, Xi'an 710000, China
| | - Xiaocao Liu
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China
| |
Collapse
|
7
|
Mecocci S, Pietrucci D, Milanesi M, Capomaccio S, Pascucci L, Evangelista C, Basiricò L, Bernabucci U, Chillemi G, Cappelli K. Comparison of colostrum and milk extracellular vesicles small RNA cargo in water buffalo. Sci Rep 2024; 14:17991. [PMID: 39097641 PMCID: PMC11297979 DOI: 10.1038/s41598-024-67249-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024] Open
Abstract
Recently, much interest has been raised for the characterization of signaling molecules carried by extracellular vesicles (EVs), which are particularly enriched in milk (mEVs). Such interest is linked to the capability of EVs to cross biological barriers, resist acidification in the gastric environment, and exert modulation of the immune system, mainly through their microRNA (miRNA) content. We characterized the small-RNA cargo of colostrum EVs (colosEVs) and mEVs from Italian Mediterranean buffalo through next generation sequencing. Colostrum (first milking after birth) and milk (day 50 of lactation) were sampled from seven subjects from five farms. ColosEVs and mEVs were subjected to morphological characterization, followed by high-depth sequencing of small RNA libraries produced from total RNA. The main difference was the amount of EV in the two samples, with colostrum showing 10 to 100-fold higher content than milk. For both matrices, miRNA was the most abundant RNA species (95% for colosEVs and 96% for mEVs) and three lists were identified: colosEV-specific, mEV-specific and shared most expressed. Gene ontology (GO) enrichment analysis on miRNA targets highlighted many terms related to the epigenetic, transcriptional and translational regulations across the three lists, with a higher number of enriched terms for colosEV-specific miRNAs. Terms specific to colosEVs were related to "cell differentiation" and "microvillus assembly", while for mEV "cardiac and blood vessel development" and "mitochondria" emergerd. Immune modulation terms were found for both sample-specific miRNAs. Overall, both matrices carry a similar molecular message in terms of biological processes potentially modulated into receiving cells, but there is significant difference in the abundance, with colostrum containing much more EVs than milk. Moreover, colosEVs carry molecules involved in signal transduction, cell cycle and immune response, as for mEVs and EVs of other previously characterized species, but with a special enrichment for miRNAs with epigenetic regulation capacities. These beneficial characteristics of colosEVs and mEVs are essential for the calf and could also be exploited for the therapeutic purposes in humans, although further studies are necessary to measure the sanitization treatment impact on EV conservation, especially in buffalo where milk is consumed almost exclusively after processing.
Collapse
Affiliation(s)
- Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, 06126, Perugia, Italy
| | - Daniele Pietrucci
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100, Viterbo, Italy
| | - Marco Milanesi
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100, Viterbo, Italy
| | - Stefano Capomaccio
- Department of Veterinary Medicine, University of Perugia, 06126, Perugia, Italy
| | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, 06126, Perugia, Italy
| | - Chiara Evangelista
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100, Viterbo, Italy
| | - Loredana Basiricò
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100, Viterbo, Italy
| | - Umberto Bernabucci
- Department of Agriculture and Forest Sciences (DAFNE), University of Tuscia, 01100, Viterbo, Italy
| | - Giovanni Chillemi
- Department for Innovation in Biological, Agro-Food and Forest Systems (DIBAF), University of Tuscia, 01100, Viterbo, Italy.
- Institute of Translational Pharmacology, National Research Council, CNR, 00133, Rome, Italy.
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, 06126, Perugia, Italy
| |
Collapse
|
8
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
9
|
Meng WT, Zhu J, Wang YC, Shao CL, Li XY, Lu PP, Huang MY, Mou FF, Guo HD, Ji G. Targeting delivery of miR-146a via IMTP modified milk exosomes exerted cardioprotective effects by inhibiting NF-κB signaling pathway after myocardial ischemia-reperfusion injury. J Nanobiotechnology 2024; 22:382. [PMID: 38951872 PMCID: PMC11218161 DOI: 10.1186/s12951-024-02631-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 06/11/2024] [Indexed: 07/03/2024] Open
Abstract
Reperfusion therapy is critical for saving heart muscle after myocardial infarction, but the process of restoring blood flow can itself exacerbate injury to the myocardium. This phenomenon is known as myocardial ischemia-reperfusion injury (MIRI), which includes oxidative stress, inflammation, and further cell death. microRNA-146a (miR-146a) is known to play a significant role in regulating the immune response and inflammation, and has been studied for its potential impact on the improvement of heart function after myocardial injury. However, the delivery of miR-146a to the heart in a specific and efficient manner remains a challenge as extracellular RNAs are unstable and rapidly degraded. Milk exosomes (MEs) have been proposed as ideal delivery platform for miRNA-based therapy as they can protect miRNAs from RNase degradation. In this study, the effects of miR-146a containing MEs (MEs-miR-146a) on improvement of cardiac function were examined in a rat model of MIRI. To enhance the targeting delivery of MEs-miR-146a to the site of myocardial injury, the ischemic myocardium-targeted peptide IMTP was modified onto the surfaces, and whether the modified MEs-miR-146a could exert a better therapeutic role was examined by echocardiography, myocardial injury indicators and the levels of inflammatory factors. Furthermore, the expressions of miR-146a mediated NF-κB signaling pathway-related proteins were detected by western blotting and qRT-PCR to further elucidate its mechanisms. MiR-146 mimics were successfully loaded into the MEs by electroporation at a square wave 1000 V voltage and 0.1 ms pulse duration. MEs-miR-146a can be up-taken by cardiomyocytes and protected the cells from oxygen glucose deprivation/reperfusion induced damage in vitro. Oral administration of MEs-miR-146a decreased myocardial tissue apoptosis and the expression of inflammatory factors and improved cardiac function after MIRI. The miR-146a level in myocardium tissues was significantly increased after the administration IMTP modified MEs-miR-146a, which was higher than that of the MEs-miR-146a group. In addition, intravenous injection of IMTP modified MEs-miR-146a enhanced the targeting to heart, improved cardiac function, reduced myocardial tissue apoptosis and suppressed inflammation after MIRI, which was more effective than the MEs-miR-146a treatment. Moreover, IMTP modified MEs-miR-146a reduced the protein levels of IRAK1, TRAF6 and p-p65. Therefore, IMTP modified MEs-miR-146a exerted their anti-inflammatory effect by inhibiting the IRAK1/TRAF6/NF-κB signaling pathway. Taken together, our findings suggested miR-146a containing MEs may be a promising strategy for the treatment of MIRI with better outcome after modification with ischemic myocardium-targeted peptide, which was expected to be applied in clinical practice in future.
Collapse
Affiliation(s)
- Wan-Ting Meng
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ya-Chao Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chang-le Shao
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiu-Ya Li
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ping-Ping Lu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Meng-Ying Huang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Fang-Fang Mou
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Hai-Dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, China-Canada Center of Research for Digestive Diseases (ccCRDD), Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
10
|
Wu T, Wang L, Gao C, Jian C, Liu Y, Fu Z, Shi C. Treg-Derived Extracellular Vesicles: Roles in Diseases and Theranostics. Mol Pharm 2024; 21:2659-2672. [PMID: 38695194 DOI: 10.1021/acs.molpharmaceut.4c00233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Regulatory T cells (Tregs), a subset of CD4+ T cells, are indispensable in maintaining immune self-tolerance and have been utilized in various diseases. Treg-derived extracellular vesicles (Treg-EVs) have been discovered to play an important role in the mechanism of Treg functions. As cell-derived membranous particles, EVs carry multiple bioactive substances that possess tremendous potential for theranostics. Treg-EVs are involved in numerous physiological and pathological processes, carrying proteins and miRNAs inherited from the parental cells. To comprehensively understand the function of Treg-EVs, here we reviewed the classification of Treg-EVs, the active molecules in Treg-EVs, their various applications in diseases, and the existing challenges for Treg-EVs based theranostics. This Review aims to clarify the feasibility and potential of Treg-EVs in diseases and theranostics, facilitating further research and application of Treg-EVs.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Jian
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Yajing Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| |
Collapse
|
11
|
Zhang S, Wang Q, Tan DEL, Sikka V, Ng CH, Xian Y, Li D, Muthiah M, Chew NWS, Storm G, Tong L, Wang J. Gut-liver axis: Potential mechanisms of action of food-derived extracellular vesicles. J Extracell Vesicles 2024; 13:e12466. [PMID: 38887165 PMCID: PMC11183959 DOI: 10.1002/jev2.12466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 05/03/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Food-derived extracellular vesicles (FEVs) are nanoscale membrane vesicles obtained from dietary materials such as breast milk, plants and probiotics. Distinct from other EVs, FEVs can survive the harsh degrading conditions in the gastrointestinal tract and reach the intestines. This unique feature allows FEVs to be promising prebiotics in health and oral nanomedicine for gut disorders, such as inflammatory bowel disease. Interestingly, therapeutic effects of FEVs have recently also been observed in non-gastrointestinal diseases. However, the mechanisms remain unclear or even mysterious. It is speculated that orally administered FEVs could enter the bloodstream, reach remote organs, and thus exert therapeutic effects therein. However, emerging evidence suggests that the amount of FEVs reaching organs beyond the gastrointestinal tract is marginal and may be insufficient to account for the significant therapeutic effects achieved regarding diseases involving remote organs such as the liver. Thus, we herein propose that FEVs primarily act locally in the intestine by modulating intestinal microenvironments such as barrier integrity and microbiota, thereby eliciting therapeutic impact remotely on the liver in non-gastrointestinal diseases via the gut-liver axis. Likewise, drugs delivered to the gastrointestinal system through FEVs may act via the gut-liver axis. As the liver is the main metabolic hub, the intestinal microenvironment may be implicated in other metabolic diseases. In fact, many patients with non-alcoholic fatty liver disease, obesity, diabetes and cardiovascular disease suffer from a leaky gut and dysbiosis. In this review, we provide an overview of the recent progress in FEVs and discuss their biomedical applications as therapeutic agents and drug delivery systems, highlighting the pivotal role of the gut-liver axis in the mechanisms of action of FEVs for the treatment of gut disorders and metabolic diseases.
Collapse
Affiliation(s)
- Sitong Zhang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Qiyue Wang
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Daniel En Liang Tan
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Vritika Sikka
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cheng Han Ng
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
| | - Yan Xian
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dan Li
- Department of Food Science and Technology, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of MedicineNational University HospitalSingaporeSingapore
- National University Centre for Organ TransplantationNational University Health SystemSingaporeSingapore
| | - Nicholas W. S. Chew
- Department of CardiologyNational University Heart CentreNational University Health SystemSingaporeSingapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Lingjun Tong
- Jinan Central HospitalShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesJinanChina
| | - Jiong‐Wei Wang
- Department of Surgery, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Cardiovascular Research Institute (CVRI)National University Heart Centre Singapore (NUHCS)SingaporeSingapore
- Department of Physiology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
12
|
K N, Bakkannavar SM, Bhat VR, Sirur FM. A review on snake venom extracellular vesicles: Past to present. Toxicon 2024; 244:107772. [PMID: 38768828 DOI: 10.1016/j.toxicon.2024.107772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Around 95% of snake venom is protein. Along with the soluble proteins, snake venom also contains proteins encapsulated in vesicles known as Snake Venom Extracellular Vesicles (SVEV). SVEVs are nano-sized membrane-bound vesicles released from the snake venom gland cells. The available published research works on SVEVs are minimal. Extracellular vesicles in the Snake Venom gland were initially discovered during the histopathological analysis of the Crotalus durissus terrificus snakes' venom gland. Later, various techniques were employed to isolate and characterize the SVEVs. The cargo of SVEV consists of a variety of proteins like Phospholipase A-2, C-type Lectins, L-Amino Acid Oxidase, Cysteine-Rich Secretory Proteins, Serine Proteinases, Dipeptidyl Peptidase-IV, Aminopeptidase-A, Ecto-5'-nucleotidases, Disintegrins. Proteomic data revealed the presence of some exclusive proteins in the SVEVs, and the other proteins are in varying concentrations in the SVEVs compared to their whole Venom. Interaction of SVEVs with mammalian cell lines showed the disruption of primary physiological functions leads to host immune modulation, and long-term effects of envenoming. Snakebite victim's blood showed variations in the specific Extracellular vesicle concentration. It has been hypothesized that SVEVs are responsible for long-term toxicity. The current review focuses on the various techniques adopted to isolate and characterize SVEVs and discusses the exclusiveness and variations of SVEV proteins and their role in snakebites.
Collapse
Affiliation(s)
- Nagendra K
- Research Scholar, Department of Forensic Medicine and Toxicology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| | - Shankar M Bakkannavar
- Associate Professor, Department of Forensic Medicine and Toxicology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| | - Vinutha R Bhat
- Associate Professor, Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| | - Freston Marc Sirur
- Associate Professor, Department of Emergency Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
13
|
Ma L, Huo Y, Tang Q, Wang X, Wang W, Wu D, Li Y, Chen L, Wang S, Zhu Y, Wang W, Liu Y, Xu N, Chen L, Yu G, Chen J. Human Breast Milk Exosomal miRNAs are Influenced by Premature Delivery and Affect Neurodevelopment. Mol Nutr Food Res 2024; 68:e2300113. [PMID: 38644336 DOI: 10.1002/mnfr.202300113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 02/27/2024] [Indexed: 04/23/2024]
Abstract
SCOPE This study investigates the exosomal microRNA (miRNA) profiles of term and preterm breast milk, including the most abundant and differentially expressed (DE) miRNAs, and their impact on neurodevelopment in infants. METHODS AND RESULTS Mature milk is collected from the mothers of term and preterm infants. Using high-throughput sequencing and subsequent data analysis, exosomal miRNA profiles of term and preterm human breast milk (HBM) are acquired and it is found that the let-7 and miR-148 families are the most abundant miRNAs. Additionally, 23 upregulated and 15 downregulated miRNAs are identified. MiR-3168 is the most upregulated miRNA in preterm HBM exosome, exhibiting targeting activity toward multiple genes involved in the SMAD and MAPK signaling pathways and playing a crucial role in early neurodevelopment. Additionally, the effects of miR-3168 on neurodevelopment is confirmed and it is determined that it is an essential factor in the differentiation of neural stem cells (NSCs). CONCLUSION This study demonstrates that miRNA expression in breast milk exosomes can be influenced by preterm delivery, thereby potentially impacting neurodevelopment in preterm infants.
Collapse
Affiliation(s)
- Ling Ma
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yanyan Huo
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Qingyuan Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiulian Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Weiqin Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Dan Wu
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yicheng Li
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Lingyan Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
- Department of Occupational Therapy Science, Nagasaki University Graduate School of Biomedical Science, 1-7-1 Sakamoto, Nagasaki, 852-8520, Japan
| | - Shasha Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yiwen Zhu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenli Wang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuan Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Nanjie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Chen
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| | - Guangjun Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinjin Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| |
Collapse
|
14
|
Shahi S, Kang T, Fonseka P. Extracellular Vesicles in Pathophysiology: A Prudent Target That Requires Careful Consideration. Cells 2024; 13:754. [PMID: 38727289 PMCID: PMC11083420 DOI: 10.3390/cells13090754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells to perform multitudes of biological functions. Owing to their significant implications in diseases, the pathophysiological role of EVs continues to be extensively studied, leading research to neglect the need to explore their role in normal physiology. Despite this, many identified physiological functions of EVs, including, but not limited to, tissue repair, early development and aging, are attributed to their modulatory role in various signaling pathways via intercellular communication. EVs are widely perceived as a potential therapeutic strategy for better prognosis, primarily through utilization as a mode of delivery vehicle. Moreover, disease-associated EVs serve as candidates for the targeted inhibition by pharmacological or genetic means. However, these attempts are often accompanied by major challenges, such as off-target effects, which may result in adverse phenotypes. This renders the clinical efficacy of EVs elusive, indicating that further understanding of the specific role of EVs in physiology may enhance their utility. This review highlights the essential role of EVs in maintaining cellular homeostasis under different physiological settings, and also discusses the various aspects that may potentially hinder the robust utility of EV-based therapeutics.
Collapse
Affiliation(s)
| | | | - Pamali Fonseka
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (S.S.); (T.K.)
| |
Collapse
|
15
|
Salehi M, Negahdari B, Mehryab F, Shekari F. Milk-Derived Extracellular Vesicles: Biomedical Applications, Current Challenges, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8304-8331. [PMID: 38587896 DOI: 10.1021/acs.jafc.3c07899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Extracellular vesicles (EVs) are nano to-micrometer-sized sacs that are released by almost all animal and plant cells and act as intercellular communicators by transferring their cargos between the source and target cells. As a safe and scalable alternative to conditioned medium-derived EVs, milk-derived EVs (miEVs) have recently gained a great deal of popularity. Numerous studies have shown that miEVs have intrinsic therapeutic actions that can treat diseases and enhance human health. Additionally, they can be used as natural drug carriers and novel classes of biomarkers. However, due to the complexity of the milk, the successful translation of miEVs from benchtop to bedside still faces several unfilled gaps, especially a lack of standardized protocols for the isolation of high-purity miEVs. In this work, by comprehensively reviewing the bovine miEVs studies, we provide an overview of current knowledge and research on miEVs while highlighting their challenges and enormous promise as a novel class of theranostics. It is hoped that this study will pave the way for clinical applications of miEVs by addressing their challenges and opportunities.
Collapse
Affiliation(s)
- Mahsa Salehi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
| | - Fatemeh Mehryab
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 14155-6153, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
16
|
Xu YR, Zhao J, Huang HY, Lin YCD, Lee TY, Huang HD, Yang Y, Wang YF. Recent insights into breast milk microRNA: their role as functional regulators. Front Nutr 2024; 11:1366435. [PMID: 38689935 PMCID: PMC11058965 DOI: 10.3389/fnut.2024.1366435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Breast milk (BM) is a primary biofluid that plays a crucial role in infant development and the regulation of the immune system. As a class of rich biomolecules in BM, microRNAs (miRNAs) are regarded as active factors contributing to infant growth and development. Surprisingly, these molecules exhibit resilience in harsh conditions, providing an opportunity for infants to absorb them. In addition, many studies have shown that miRNAs in breast milk, when absorbed into the gastrointestinal system, can act as a class of functional regulators to effectively regulate gene expression. Understanding the absorption pattern of BM miRNA may facilitate the creation of formula with a more optimal miRNA balance and pave the way for novel drug delivery techniques. In this review, we initially present evidence of BM miRNA absorption. Subsequently, we compile studies that integrate both in vivo and in vitro findings to illustrate the bioavailability and biodistribution of BM miRNAs post-absorption. In addition, we evaluate the strengths and weaknesses of previous studies and discuss potential variables contributing to discrepancies in their outcomes. This literature review indicates that miRNAs can be absorbed and act as regulatory agents.
Collapse
Affiliation(s)
- Yi-Ran Xu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Jinglu Zhao
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Hsi-Yuan Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yang-Chi-Dung Lin
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology and Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yi Yang
- Department of Nephrology, Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu, China
| | - Yong-Fei Wang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
17
|
Luo M, Sun J, Li S, Wei L, Sun R, Feng X, Zhang H, Chen T, Xi Q, Zhang Y, Qi Q. Protective effect of Enterococcus faecium against ethanol-induced gastric injury via extracellular vesicles. Microbiol Spectr 2024; 12:e0389423. [PMID: 38488394 PMCID: PMC10986489 DOI: 10.1128/spectrum.03894-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/04/2024] [Indexed: 04/06/2024] Open
Abstract
Recently, Enterococcus has been shown to have gastric protective functions, and the mechanisms by which Enterococcus modulates gastric function are still being investigated. Herein, we investigated how Enterococcus faecium (Efm) and E. faecium-derived extracellular vesicles (EVs) (EfmEVs) exert protective effect against ethanol-induced gastric injury by investigating the effect of EfmEVs on gastric mucosal ulcer scoring, histological lesion, mucosal glycoprotein production, acidity, anti-oxidative function, and inflammatory responses in rat. Pretreatment with Efm showed significant reduction of ethanol-induced gastric injury, as evidenced by the lowering of ulcer index, histological lesion, gastric pH, and inflammatory responses and the enhancement of mucosal glycoprotein production and anti-oxidative function. Further functional studies on three bioactive components [inactivated Efm, EfmEVs (EVs), and EV-free supernatants] of the bacterial culture showed that EVs are mostly responsible for the gastroprotective effect. Moreover, EV secretion is beneficial for the gastroprotective effect of Efm. Hence, EVs mediated the protective effect of Efm against ethanol-induced gastric injury by lowering inflammatory responses and enhancing anti-oxidative function and may be a potent anti-inflammatory and anti-oxidative strategy to alleviate hyperinflammatory gastrointestinal tract conditions.IMPORTANCEThis study indicated that Enterococcus faecium provided a protective effect against rat gastric injury, which involved improvement of the mucosal glycoprotein production, anti-oxidative function, and inflammatory responses. Furthermore, we confirmed that three bioactive components (inactivated Efm, extracellular vesicles, and EV-free supernatants) of E. faecium culture also contributed to the gastroprotective effect. Importantly, E. faecium-derived EVs showed an effective impact for the gastroprotective effect.
Collapse
Affiliation(s)
- Meiying Luo
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Junhang Sun
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Suqian Li
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Limin Wei
- Sanya Institute, Hainan Academy of Agricultural Sciences (Hainan Experimental Animal Research Center), Sanya, China
| | - Ruiping Sun
- Institute of Animal Science and Veterinary Medicine of Hainan Academy of Agricultural Sciences, Haikou, China
| | - Xin Feng
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Huihua Zhang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
18
|
Saavedra LPJ, Piovan S, Moreira VM, Gonçalves GD, Ferreira ARO, Ribeiro MVG, Peres MNC, Almeida DL, Raposo SR, da Silva MC, Barbosa LF, de Freitas Mathias PC. Epigenetic programming for obesity and noncommunicable disease: From womb to tomb. Rev Endocr Metab Disord 2024; 25:309-324. [PMID: 38040983 DOI: 10.1007/s11154-023-09854-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Several epidemiological, clinical and experimental studies in recent decades have shown the relationship between exposure to stressors during development and health outcomes later in life. The characterization of these susceptible phases, such as preconception, gestation, lactation and adolescence, and the understanding of factors that influence the risk of an adult individual for developing obesity, metabolic and cardiovascular diseases, is the focus of the DOHaD (Developmental Origins of Health and Disease) research line. In this sense, advancements in molecular biology techniques have contributed significantly to the understanding of the mechanisms underlying the observed phenotypes, their morphological and physiological alterations, having as a main driving factor the epigenetic modifications and their consequent modulation of gene expression. The present narrative review aimed to characterize the different susceptible phases of development and associated epigenetic modifications, and their implication in the development of non-communicable diseases. Additionally, we provide useful insights into interventions during development to counteract or prevent long-term programming for disease susceptibility.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maria Natália Chimirri Peres
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Scarlett Rodrigues Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Mariane Carneiro da Silva
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Letícia Ferreira Barbosa
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil.
| |
Collapse
|
19
|
Li T, Chen X, Qi Q, Feng X. Bovine Milk Derived Exosomes Affect Gut Microbiota of DSS-Induced Colitis Mice. Indian J Microbiol 2024; 64:100-109. [PMID: 38468747 PMCID: PMC10924850 DOI: 10.1007/s12088-023-01131-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/27/2023] [Indexed: 03/13/2024] Open
Abstract
The objective of this study was to investigate the effect of bovine milk derived exosomes (MDEs) on the gut microbiota of Dextran sodium sulfate (DSS)-induced colitis mice. Total of 42 specific pathogen free (SPF) male BALB/c mice (3 weeks old) were randomly assigned to three groups including control group, DSS group (DSS) and bovine milk derived exosome group (Exo), with 7 replicates/cages per treatment and two mice in one cage. 16S rRNA gene sequencing of cecal digesta samples was conducted. DSS significantly decreased the average daily feed intake of mice in DSS and Exo groups (P = 0.03). Shannon index of the DSS group was significantly lower than the control group (P < 0.05) whereas no difference between the control group and Exo group was observed. Administration of MDEs tended to increase the relative abundance of Campylobaterota. Compared to the control group, the relative abundance of Roseburia was significantly decreased in the DSS group (P < 0.05) whereas no difference between the Exo group and control group was observed. MDEs also tended to increase the relative abundance of Lachnospiraceae_UCG_006. In conclusion, oral administration of 10 µL MDEs (1 mg/mL) positively affected gut microbiota of DSS-induced colitis mice. The results of this study provided valuable reference for MDEs application in the prevention and treatment of colitis.
Collapse
Affiliation(s)
- Tonghao Li
- School of Life Science and Engineering, Foshan University, No. 33 Guangyun Road, Nanhai District, Foshan, Guangdong China
| | - Xiaolin Chen
- School of Life Science and Engineering, Foshan University, No. 33 Guangyun Road, Nanhai District, Foshan, Guangdong China
| | - Qien Qi
- School of Life Science and Engineering, Foshan University, No. 33 Guangyun Road, Nanhai District, Foshan, Guangdong China
| | - Xin Feng
- School of Life Science and Engineering, Foshan University, No. 33 Guangyun Road, Nanhai District, Foshan, Guangdong China
| |
Collapse
|
20
|
Surzenko N, Bastidas J, Reid RW, Curaba J, Zhang W, Bostan H, Wilson M, Dominique A, Roberson J, Ignacio G, Komarnytsky S, Sanders A, Lambirth K, Brouwer CR, El-Khodor BF. Functional recovery following traumatic brain injury in rats is enhanced by oral supplementation with bovine thymus extract. FASEB J 2024; 38:e23460. [PMID: 38315443 DOI: 10.1096/fj.202301859r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/30/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death worldwide. There are currently no effective treatments for TBI, and trauma survivors suffer from a variety of long-lasting health consequences. With nutritional support recently emerging as a vital step in improving TBI patients' outcomes, we sought to evaluate the potential therapeutic benefits of nutritional supplements derived from bovine thymus gland, which can deliver a variety of nutrients and bioactive molecules. In a rat model of controlled cortical impact (CCI), we determined that animals supplemented with a nuclear fraction of bovine thymus (TNF) display greatly improved performance on beam balance and spatial memory tests following CCI. Using RNA-Seq, we identified an array of signaling pathways that are modulated by TNF supplementation in rat hippocampus, including those involved in the process of autophagy. We further show that bovine thymus-derived extracts contain antigens found in neural tissues and that supplementation of rats with thymus extracts induces production of serum IgG antibodies against neuronal and glial antigens, which may explain the enhanced animal recovery following CCI through possible oral tolerance mechanism. Collectively, our data demonstrate, for the first time, the potency of a nutritional supplement containing nuclear fraction of bovine thymus in enhancing the functional recovery from TBI.
Collapse
Affiliation(s)
- Natalia Surzenko
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | | | - Robert W Reid
- College of Computing and Informatics, University of North Carolina at Charlotte, Kannapolis, North Carolina, USA
| | - Julien Curaba
- Eremid Genomic Services, LLC, Kannapolis, North Carolina, USA
| | - Wei Zhang
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | - Hamed Bostan
- Eremid Genomic Services, LLC, Kannapolis, North Carolina, USA
| | - Mickey Wilson
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | - Ashley Dominique
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | - Julia Roberson
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| | - Glicerio Ignacio
- David H. Murdock Research Institute, Kannapolis, North Carolina, USA
| | - Slavko Komarnytsky
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, North Carolina State University, Kannapolis, North Carolina, USA
| | - Alexa Sanders
- College of Computing and Informatics, University of North Carolina at Charlotte, Kannapolis, North Carolina, USA
| | - Kevin Lambirth
- College of Computing and Informatics, University of North Carolina at Charlotte, Kannapolis, North Carolina, USA
| | - Cory R Brouwer
- College of Computing and Informatics, University of North Carolina at Charlotte, Kannapolis, North Carolina, USA
| | - Bassem F El-Khodor
- Nutrition Innovation Center, Standard Process, Inc., Kannapolis, North Carolina, USA
| |
Collapse
|
21
|
Yung C, Zhang Y, Kuhn M, Armstrong RJ, Olyaei A, Aloia M, Scottoline B, Andres SF. Neonatal enteroids absorb extracellular vesicles from human milk-fed infant digestive fluid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.03.556067. [PMID: 38187651 PMCID: PMC10769189 DOI: 10.1101/2023.09.03.556067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Human milk contains extracellular vesicles (HMEVs). Pre-clinical models suggest that HMEVs may enhance intestinal function and limit inflammation; however, it is unknown if HMEVs or their cargo survive neonatal human digestion. This limits the ability to leverage HMEV cargo as additives to infant nutrition or as therapeutics. This study aimed to develop an EV isolation pipeline from small volumes of human milk and neonatal intestinal contents after milk feeding (digesta) to address the hypothesis that HMEVs survive in vivo neonatal digestion to be taken up intestinal epithelial cells (IECs). Digesta was collected from nasoduodenal sampling tubes or ostomies. EVs were isolated from raw and pasteurized human milk and digesta by density-gradient ultracentrifugation following two-step skimming, acid precipitation of caseins, and multi-step filtration. EVs were validated by electron microscopy, western blotting, nanoparticle tracking analysis, resistive pulse sensing, and super-resolution microscopy. EV uptake was tested in human neonatal enteroids. HMEVs and digesta EVs (dEVs) show typical EV morphology and are enriched in CD81 and CD9, but depleted of β-casein and lactalbumin. HMEV and some dEV fractions contain mammary gland-derived protein BTN1A1. Neonatal human enteroids rapidly take up dEVs in part via clathrin-mediated endocytosis. Our data suggest that EVs can be isolated from digestive fluid and that these dEVs can be absorbed by IECs.
Collapse
|
22
|
Doerfler R, Yerneni S, Newby A, Chaudhary N, Shu A, Fein K, Hofstatter Azambuja J, Whitehead KA. Characterization and comparison of human and mouse milk cells. PLoS One 2024; 19:e0297821. [PMID: 38295101 PMCID: PMC10830055 DOI: 10.1371/journal.pone.0297821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024] Open
Abstract
Recent data has characterized human milk cells with unprecedented detail and provided insight into cell populations. While such analysis of freshly expressed human milk has been possible, studies of cell functionality within the infant have been limited to animal models. One commonly used animal model for milk research is the mouse; however, limited data are available describing the composition of mouse milk. In particular, the maternal cells of mouse milk have not been previously characterized in detail, in part due to the difficulty in collecting sufficient volumes of mouse milk. In this study, we have established a method to collect high volumes of mouse milk, isolate cells, and compare the cell counts and types to human milk. Surprisingly, we found that mouse milk cell density is three orders of magnitude higher than human milk. The cell types present in the milk of mice and humans are similar, broadly consisting of mammary epithelial cells and immune cells. These results provide a basis of comparison for mouse and human milk cells and will inform the most appropriate uses of mouse models for the study of human phenomena.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Saigopalakrishna Yerneni
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Alexandra Newby
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Ashley Shu
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Katherine Fein
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Juliana Hofstatter Azambuja
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kathryn A. Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| |
Collapse
|
23
|
Muse ME, Armstrong DA, Hoen AG, Gilbert-Diamond D, Gui J, Palys TJ, Kolling FW, Christensen BC, Karagas MR, Howe CG. Maternal-Infant Factors in Relation to Extracellular Vesicle and Particle miRNA in Prenatal Plasma and in Postpartum Human Milk. Int J Mol Sci 2024; 25:1538. [PMID: 38338815 PMCID: PMC10855220 DOI: 10.3390/ijms25031538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
MicroRNAs (miRNA) in extracellular vesicles and particles (EVPs) in maternal circulation during pregnancy and in human milk postpartum are hypothesized to facilitate maternal-offspring communication via epigenetic regulation. However, factors influencing maternal EVP miRNA profiles during these two critical developmental windows remain largely unknown. In a pilot study of 54 mother-child dyads in the New Hampshire Birth Cohort Study, we profiled 798 EVP miRNAs, using the NanoString nCounter platform, in paired maternal second-trimester plasma and mature (6-week) milk samples. In adjusted models, total EVP miRNA counts were lower for plasma samples collected in the afternoon compared with the morning (p = 0.024). Infant age at sample collection was inversely associated with total miRNA counts in human milk EVPs (p = 0.040). Milk EVP miRNA counts were also lower among participants who were multiparous after delivery (p = 0.047), had a pre-pregnancy BMI > 25 kg/m2 (p = 0.037), or delivered their baby via cesarean section (p = 0.021). In post hoc analyses, we also identified 22 specific EVP miRNA that were lower among participants who delivered their baby via cesarean section (Q < 0.05). Target genes of delivery mode-associated miRNAs were over-represented in pathways related to satiety signaling in infants (e.g., CCKR signaling) and mammary gland development and lactation (e.g., FGF signaling, EGF receptor signaling). In conclusion, we identified several key factors that may influence maternal EVP miRNA composition during two critical developmental windows, which should be considered in future studies investigating EVP miRNA roles in maternal and child health.
Collapse
Affiliation(s)
- Meghan E. Muse
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - David A. Armstrong
- Research Service, V.A. Medical Center, Hartford, VT 05009, USA
- Department of Dermatology, Dartmouth Health, Lebanon, NH 03756, USA
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Diane Gilbert-Diamond
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Jiang Gui
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Thomas J. Palys
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Frederick W. Kolling
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Brock C. Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Caitlin G. Howe
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| |
Collapse
|
24
|
Marquez CA, Oh CI, Ahn G, Shin WR, Kim YH, Ahn JY. Synergistic vesicle-vector systems for targeted delivery. J Nanobiotechnology 2024; 22:6. [PMID: 38167116 PMCID: PMC10763086 DOI: 10.1186/s12951-023-02275-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
With the immense progress in drug delivery systems (DDS) and the rise of nanotechnology, challenges such as target specificity remain. The vesicle-vector system (VVS) is a delivery system that uses lipid-based vesicles as vectors for a targeted drug delivery. When modified with target-probing materials, these vesicles become powerful vectors for drug delivery with high target specificity. In this review, we discuss three general types of VVS based on different modification strategies: (1) vesicle-probes; (2) vesicle-vesicles; and (3) genetically engineered vesicles. The synthesis of each VVS type and their corresponding properties that are advantageous for targeted drug delivery, are also highlighted. The applications, challenges, and limitations of VVS are briefly examined. Finally, we share a number of insights and perspectives regarding the future of VVS as a targeted drug delivery system at the nanoscale.
Collapse
Affiliation(s)
- Christine Ardelle Marquez
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Cho-Im Oh
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
| | - Gna Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Woo-Ri Shin
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea
- Department of Bioengineering, University of Pennsylvania, 210 S 33rd St, Philadelphia, PA, 19104, USA
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Ji-Young Ahn
- Department of Microbiology, Chungbuk National University, 1 Chungdae-Ro, Seowon-Gu, Cheongju, 28644, Republic of Korea.
- Center for Ecology and Environmental Toxicology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
25
|
Guo ZY, Tang Y, Cheng YC. Exosomes as Targeted Delivery Drug System: Advances in Exosome Loading, Surface Functionalization and Potential for Clinical Application. Curr Drug Deliv 2024; 21:473-487. [PMID: 35702803 DOI: 10.2174/1567201819666220613150814] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022]
Abstract
Exosomes are subtypes of vesicles secreted by almost all cells and can play an important role in intercellular communication. They contain various proteins, lipids, nucleic acids and other natural substances from their metrocytes. Exosomes are expected to be a new generation of drug delivery systems due to their low immunogenicity, high potential to transfer bioactive substances and biocompatibility. However, exosomes themselves are not highly targeted, it is necessary to develop new surface modification techniques and targeted drug delivery strategies, which are the focus of drug delivery research. In this review, we introduced the biogenesis of exosomes and their role in intercellular communication. We listed various advanced exosome drug-loading techniques. Emphatically, we summarized different exosome surface modification techniques and targeted drug delivery strategies. In addition, we discussed the application of exosomes in vaccines and briefly introduced milk exosomes. Finally, we clarified the clinical application prospects and shortcomings of exosomes.
Collapse
Affiliation(s)
- Zun Y Guo
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| | - Yue Tang
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| | - Yi C Cheng
- Department of Pharmacy, China Pharmaceutical University, No.639, Longmian Avenue, Nanjing 211198, P.R. China
| |
Collapse
|
26
|
Vahkal B, Altosaar I, Tremblay E, Gagné D, Hüttman N, Minic Z, Côté M, Blais A, Beaulieu J, Ferretti E. Gestational age at birth influences protein and RNA content in human milk extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e128. [PMID: 38938674 PMCID: PMC11080785 DOI: 10.1002/jex2.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 06/29/2024]
Abstract
Human milk extracellular vesicles (HM EVs) are proposed to protect against disease development in infants. This protection could in part be facilitated by the bioactive EV cargo of proteins and RNA. Notably, mothers birth infants of different gestational ages with unique needs, wherein the EV cargo of HM may diverge. We collected HM from lactating mothers within two weeks of a term or preterm birth. Following purification of EVs, proteins and mRNA were extracted for proteomics and sequencing analyses, respectively. Over 2000 protein groups were identified, and over 8000 genes were quantified. The total number of proteins and mRNA did not differ significantly between the two conditions, while functional bioinformatics of differentially expressed cargo indicated enrichment in immunoregulatory cargo for preterm HM EVs. In term HM EVs, significantly upregulated cargo was enriched in metabolism-related functions. Based on gene expression signatures from HM-contained single cell sequencing data, we proposed that a larger portion of preterm HM EVs are secreted by immune cells, whereas term HM EVs contain more signatures of lactocyte epithelial cells. Proposed differences in EV cargo could indicate variation in mother's milk based on infants' gestational age and provide basis for further functional characterisation.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Illimar Altosaar
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
| | - Eric Tremblay
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - David Gagné
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - Nico Hüttman
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Zoran Minic
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
- Brain and Mind InstituteUniversity of OttawaOttawaCanada
- Éric Poulin Centre for Neuromuscular DiseaseOttawaCanada
| | | | - Emanuela Ferretti
- Department of Pediatrics, Division of NeonatologyChildren's Hospital of Eastern OntarioOttawaCanada
| |
Collapse
|
27
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
28
|
Ávila Morales G, De Leonardis D, Filipe J, Furioso Ferreira R, Agazzi A, Sauerwein H, Comi M, Mrljak V, Lecchi C, Ceciliani F. Porcine milk exosomes modulate the immune functions of CD14 + monocytes in vitro. Sci Rep 2023; 13:21447. [PMID: 38052991 PMCID: PMC10698175 DOI: 10.1038/s41598-023-48376-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/25/2023] [Indexed: 12/07/2023] Open
Abstract
Exosomes mediate near and long-distance intercellular communication by transferring their molecular cargo to recipient cells, altering their biological response. Milk exosomes (MEx) are internalized by immune cells and exert immunomodulatory functions in vitro. Porcine MEx can accumulate in the small intestine, rich in macrophages. No information is available on the immunomodulatory ability of porcine MEx on porcine monocytes, which are known precursors of gut macrophages. Therefore, this study aims at (1) assessing the in vitro uptake of porcine MEx by porcine monocytes (CD14+), and (2) evaluating the in vitro impact of porcine MEx on porcine monocytes immune functions. MEx were purified by ultracentrifugation and size exclusion chromatography. The monocytes' internalization of PKH26-labeled MEx was examined using fluorescence microscopy. Monocytes were incubated with increasing exosome concentrations and their apoptosis and viability were measured. Lastly, the ability of MEx to modulate the cells' immune activities was evaluated by measuring monocytes' phagocytosis, the capacity of killing bacteria, chemotaxis, and reactive oxygen species (ROS) production. MEx were internalized by porcine monocytes in vitro. They also decreased their chemotaxis and phagocytosis, and increased ROS production. Altogether, this study provides insights into the role that MEx might play in pigs' immunity by demonstrating that MEx are internalized by porcine monocytes in vitro and exert immunomodulatory effects on inflammatory functions.
Collapse
Affiliation(s)
- Gabriela Ávila Morales
- Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Lodi, Italy.
| | - Daria De Leonardis
- Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Lodi, Italy
| | - Joel Filipe
- Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Lodi, Italy
| | - Rafaela Furioso Ferreira
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Alessandro Agazzi
- Department of Veterinary Science for Health, Animal Production and Alimentary Security, Università Degli Studi di Milano, Lodi, Italy
| | - Helga Sauerwein
- Institute of Animal Science, Physiology and Hygiene Unit, University of Bonn, Bonn, Germany
| | - Marcello Comi
- Department of Human Science and Quality of Life Promotion, Università Telematica San Raffaele, Rome, Italy
| | - Vladimir Mrljak
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Cristina Lecchi
- Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Lodi, Italy
| | - Fabrizio Ceciliani
- Department of Veterinary Medicine and Animal Sciences, Università Degli Studi di Milano, Lodi, Italy
| |
Collapse
|
29
|
Yeruva L, Mulakala BK, Rajasundaram D, Gonzalez S, Cabrera-Rubio R, Martínez-Costa C, Collado MC. Human milk miRNAs associate to maternal dietary nutrients, milk microbiota, infant gut microbiota and growth. Clin Nutr 2023; 42:2528-2539. [PMID: 37931372 DOI: 10.1016/j.clnu.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Maternal diet influences the milk composition, yet little information is available on the impact of maternal diet on milk miRNAs expression. Further, the association of human milk miRNAs to maternal diet and milk microbiota is not explored. In addition, the role of milk miRNAs on the infant gut microbiota, infant growth and development has not been investigated. METHODS Milk samples were collected from 60 healthy lactating women at ≤15d post-partum, HTG transcriptome assay was performed to examine milk miRNA profile. Maternal clinical and dietary clusters information were available and infant anthropometric measures were followed up to one year of age. Milk and infant microbiota were analyzed by 16S rRNA gene sequencing and integrative multi-omics data analysis was performed to identify potential association between microRNA, maternal dietary nutrients and microbiota. RESULTS Discriminant analysis revealed that the milk miRNAs were clustered into groups according to the maternal protein source. Interestingly, 31 miRNAs were differentially expressed (P adj < 0.05) between maternal dietary clusters (Cluster 1: enriched in plant protein and fibers and Cluster 2: enriched in animal protein), with 30 miRNAs downregulated in the plant protein group relative to animal protein group. Pathway analysis revealed that the top enriched pathways (P adj < 0.01) were involved in cell growth and proliferation processes. Furthermore, significant features contributing to the clustering were associated with maternal dietary nutrients and milk microbiota (r > 0.70). Further, miR-378 and 320 family miRNAs involved in adipogenesis were positively correlated to the infant BMI-z-scores, weight, and weight for length-z-scores at 6 months of age. CONCLUSIONS Maternal dietary source impacts the milk miRNA expression profile. Further, miRNAs were associated with maternal dietary nutrients, milk microbiota and to the infant gut microbiota and infant growth and development. CLINICAL TRIAL The study is registered in ClinicalTrials.gov. The identification number is NCT03552939.
Collapse
Affiliation(s)
- Laxmi Yeruva
- Microbiome and Metabolism Research Unit, USDA-ARS, SEA, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA.
| | - Bharat Kumar Mulakala
- Microbiome and Metabolism Research Unit, USDA-ARS, SEA, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA; Texas A&M AgriLife Institute for Advancing Health Through Agriculture, TX, USA
| | | | - Sonia Gonzalez
- Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA, ISPA), Oviedo, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
30
|
Sukreet S, Braga CP, Adamec J, Cui J, Zempleni J. The absorption of bovine milk small extracellular vesicles largely depends on galectin 3 and galactose ligands in human intestinal cells and C57BL/6J mice. Am J Physiol Cell Physiol 2023; 325:C1421-C1430. [PMID: 37955122 PMCID: PMC10861145 DOI: 10.1152/ajpcell.00282.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023]
Abstract
Small extracellular vesicles in milk (sMEVs) have attracted attention in drug delivery and as bioactive food compounds. Previous studies implicate galactose residues on the sMEV surface in sMEV transport across intestinal and endothelial barriers in humans, but details of glycoprotein-dependent transport are unknown. We used a combination of cell biology and genetics protocols to identify glycoproteins on the sMEV surface that facilitate sMEV absorption. We identified 256 proteins on the bovine sMEVs surface by using LC-MS/MS, and bioinformatics analysis suggested that 42, 13, and 13 surface proteins were N-, O-, and 13 C-glycosylated, respectively. Lectin blots confirmed the presence of mannose, galactose, N-acetyl galactose, fucose, and neuraminate. When surface proteins were removed by various treatment with various proteases, sMEV uptake decreased by up to 58% and 67% in FHs-74 Int and Caco-2 cells, respectively, compared with controls (P < 0.05). When glycans were removed by treatment with various glycosidases, sMEV uptake decreased by up to 54% and 74% in FHs-74 Int and Caco-2 cells, respectively (P < 0.05). When galactose and N-acetyl galactosamine residues were blocked with agglutinins, sMEV uptake decreased by more than 50% in FHs-74 Int cells (P < 0.05). When bovine sMEVs were administered to Galectin-3 knockout mice by oral gavage, hepatic sMEV accumulation decreased by 56% compared with wild-type mice (P < 0.05), consistent with a role of β-galactoside glycan structures in the absorption of sMEVs. We conclude that sMEVs are decorated with glycoproteins, and Galectin-3 and its galactose ligands are particularly important for sMEV absorption.NEW & NOTEWORTHY This is the first paper to assess the role of unique glycans and their Galectin-3 receptor in the transport and distribution of small extracellular vesicles ("exosomes") from milk in mammals. The research assessed milk exosome transport and distribution by using multiple approaches and platforms including cell cultures, various exosome labels, knockout and mutant mice, enzymatic removal of surface proteins and glycans, and lectin blocking of glycans.
Collapse
Affiliation(s)
- Sonal Sukreet
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Camila Pereira Braga
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Jiri Adamec
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Juan Cui
- School of Computing, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States
| |
Collapse
|
31
|
Słyk-Gulewska P, Kondracka A, Kwaśniewska A. MicroRNA as a new bioactive component in breast milk. Noncoding RNA Res 2023; 8:520-526. [PMID: 37520770 PMCID: PMC10371784 DOI: 10.1016/j.ncrna.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/24/2023] [Accepted: 06/25/2023] [Indexed: 08/01/2023] Open
Abstract
Breast milk is a complex and multifaceted fluid that plays a critical role in the development of infants. It is composed of water, carbohydrates, fats, proteins, vitamins, and minerals, as well as numerous bioactive compounds such as hormones, oligosaccharides, and immune proteins. Additionally, breast milk contains microRNAs, which have been found to regulate gene expression and impact various aspects of infant development. This text provides an overview of the components of human breast milk and their importance in infant development, with a focus on microRNAs. MicroRNAs are short RNA sequences that regulate gene expression posttranscriptionally, and they play an important role in shaping the mechanisms of immunity, protecting against oxidative stress, and promoting thermogenesis. The composition of breast milk can vary in the same mother between different feedings, as it changes in response to various factors such as the infant's age, feeding frequency and duration, time of day, and maternal health status. Despite the variations in breast milk composition, it still provides complete nutrition for the infant. The unique microRNA profiles in breast milk and how they are affected by various factors can have significant implications for disease prevention and treatment. Further research is needed to better understand the functions of individual microRNA molecules and their potential therapeutic applications.
Collapse
|
32
|
Kaeffer B. Human Breast Milk miRNAs: Their Diversity and Potential for Preventive Strategies in Nutritional Therapy. Int J Mol Sci 2023; 24:16106. [PMID: 38003296 PMCID: PMC10671413 DOI: 10.3390/ijms242216106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The endogenous miRNAs of breast milk are the products of more than 1000 nonprotein-coding genes, giving rise to mature small regulatory molecules of 19-25 nucleotides. They are incorporated in macromolecular complexes, loaded on Argonaute proteins, sequestrated in exosomes and lipid complexes, or present in exfoliated cells of epithelial, endothelial, or immune origins. Their expression is dependent on the stage of lactation; however, their detection depends on progress in RNA sequencing and the reappraisal of the definition of small RNAs. Some miRNAs from plants are detected in breast milk, opening the possibility of the stimulation of immune cells from the allergy repertoire. Each miRNA harbors a seeding sequence, which targets mRNAs, gene promoters, or long noncoding RNAs. Their activities depend on their bioavailability. Efficient doses of miRNAs are estimated to be roughly 100 molecules in the cytoplasm of target cells from in vitro and in vivo experiments. Each miRNA is included in networks of stimulation/inhibition/sequestration, driving the expression of cellular phenotypes. Three types of stress applied during lactation to manipulate miRNA supply were explored using rodent offspring: a foster mother, a cafeteria diet, and early weaning. This review presents the main mature miRNAs described from current mothers' cohorts and their bioavailability in experimental models as well as studies assessing the potential of miR-26 or miR-320 miRNA families to alter offspring phenotypes.
Collapse
Affiliation(s)
- Bertrand Kaeffer
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
| |
Collapse
|
33
|
Francese R, Peila C, Donalisio M, Lamberti C, Cirrincione S, Colombi N, Tonetto P, Cavallarin L, Bertino E, Moro GE, Coscia A, Lembo D. Viruses and Human Milk: Transmission or Protection? Adv Nutr 2023; 14:1389-1415. [PMID: 37604306 PMCID: PMC10721544 DOI: 10.1016/j.advnut.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/14/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023] Open
Abstract
Human milk (HM) is considered the best source of nutrition for infant growth and health. This nourishment is unique and changes constantly during lactation to adapt to the physiological needs of the developing infant. It is also recognized as a potential route of transmission of some viral pathogens although the presence of a virus in HM rarely leads to a disease in an infant. This intriguing paradox can be explained by considering the intrinsic antiviral properties of HM. In this comprehensive and schematically presented review, we have described what viruses have been detected in HM so far and what their potential transmission risk through breastfeeding is. We have provided a description of all the antiviral compounds of HM, along with an analysis of their demonstrated and hypothesized mechanisms of action. Finally, we have also analyzed the impact of HM pasteurization and storage methods on the detection and transmission of viruses, and on the antiviral compounds of HM. We have highlighted that there is currently a deep knowledge on the potential transmission of viral pathogens through breastfeeding and on the antiviral properties of HM. The current evidence suggests that, in most cases, it is unnecessarily to deprive an infant of this high-quality nourishment and that the continuation of breastfeeding is in the best interest of the infant and the mother.
Collapse
Affiliation(s)
- Rachele Francese
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy
| | - Chiara Peila
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Manuela Donalisio
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy
| | - Cristina Lamberti
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Simona Cirrincione
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Nicoletta Colombi
- Biblioteca Federata di Medicina "Ferdinando Rossi", University of Turin, Turin, Italy
| | - Paola Tonetto
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Laura Cavallarin
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Enrico Bertino
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Guido E Moro
- Italian Association of Human Milk Banks (AIBLUD), Milan, Italy.
| | - Alessandra Coscia
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy.
| | - David Lembo
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy.
| |
Collapse
|
34
|
Zhang Y, Lin Y, He J, Song S, Luo Y, Lu Y, Chen S, Wang Q, Li Y, Ren F, Guo H. Milk-derived small extracellular vesicles: a new perspective on dairy nutrition. Crit Rev Food Sci Nutr 2023:1-22. [PMID: 37819268 DOI: 10.1080/10408398.2023.2263573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Milk contains bioactive compounds that have multiple essential benefits. Milk-derived small extracellular vesicles (M-sEVs) have emerged as novel bioactive milk components with various beneficial biological functions and broad applications. The M-sEVs from different mammalian sources have similar composition and bioactive functions. The digestive stability and biocompatibility of the M-sEVs provide a good foundation for their physiological functions. Evidence suggests that M-sEVs promote intestinal, immune, bone, neural, liver, and heart health and show therapeutic effects against cancer, indicating their potential for use in functional foods. In addition, M-sEVs can be developed as natural delivery carriers owing to their superior structural characteristics. Further studies are needed to elucidate the relationship between the specific components and functions of M-sEVs, standardize their extraction processes, and refine relevant clinical trials to advance the future applications of M-sEVs. This review summarizes the structure and composition of M-sEVs isolated from different milk sources and discusses several common extraction methods. Since the introduction of M-sEVs for digestion and absorption, studies have been conducted on their biological functions. Furthermore, we outline the theoretical industrial production route, potential application scenarios of M-sEVs, and the future perspectives of M-sEV research.
Collapse
Affiliation(s)
- Yuning Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Sijia Song
- Food Laboratory of Zhongyuan, Luohe, PR China
| | - Yujia Luo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | | | - Qingyu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
35
|
Zhang JY, Ren CQ, Cao YN, Ren Y, Zou L, Zhou C, Peng LX. Role of MicroRNAs in Dietary Interventions for Obesity and Obesity-Related Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14396-14412. [PMID: 37782460 DOI: 10.1021/acs.jafc.3c03042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Obesity and related metabolic syndromes pose a serious threat to human health and quality of life. A proper diet is a safe and effective strategy to prevent and control obesity, thus maintaining overall health. However, no consensus exists on the connotations of proper diet, and it is attributed to various factors, including "nutritional dark matter" and the "matrix effect" of food. Accumulating evidence confirms that obesity is associated with the in vivo levels of miRNAs, which serve as potential markers and regulatory targets for obesity onset and progression; food-derived miRNAs can regulate host obesity by targeting the related genes or gut microbiota across the animal kingdom. Host miRNAs mediate food nutrient-gut microbiota-obesity interactions. Thus, miRNAs are important correlates of diet and obesity onset. This review outlines the recent findings on miRNA-mediated food interventions for obesity, thereby elucidating their potential applications. Overall, we provide new perspectives and views on the evaluation of dietary nutrition, which may bear important implications for dietary control and obesity prevention.
Collapse
Affiliation(s)
- Ji-Yue Zhang
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Chao-Qin Ren
- Aba Teachers University, Wenchuan, Sichuan 623002, People's Republic of China
| | - Ya-Nan Cao
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Yuanhang Ren
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Chuang Zhou
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| | - Lian-Xin Peng
- Key Laboratory of Coarse Cereal Processing of Ministry of Agriculture and Rural Affairs, Sichuan Province Engineering Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, People's Republic of China
| |
Collapse
|
36
|
Fayyazpour P, Fayyazpour A, Abbasi K, Vaez-Gharamaleki Y, Zangbar MSS, Raeisi M, Mehdizadeh A. The role of exosomes in cancer biology by shedding light on their lipid contents. Pathol Res Pract 2023; 250:154813. [PMID: 37769395 DOI: 10.1016/j.prp.2023.154813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/08/2023] [Indexed: 09/30/2023]
Abstract
Exosomes are extracellular bilayer membrane nanovesicles released by cells after the fusion of multivesicular bodies (MVBs) with the plasma membrane. One of the interesting features of exosomes is their ability to carry and transfer various molecules, including lipids, proteins, nucleic acids, and therapeutic cargoes among cells. As intercellular signaling organelles, exosomes participate in various signaling processes such as tumor growth, metastasis, angiogenesis, epithelial-to-mesenchymal transition (EMT), and cell physiology such as cell-to-cell communication. Moreover, these particles are considered good vehicles to shuttle vaccines and drugs for therapeutic applications regarding cancers and tumor cells. These bioactive vesicles are also rich in various lipid molecules such as cholesterol, sphingomyelin (SM), glycosphingolipids, and phosphatidylserine (PS). These lipids play an important role in the formation, release, and function of the exosomes and interestingly, some lipids are used as biomarkers in cancer diagnosis. This review aimed to focus on exosomes lipid content and their role in cancer biology.
Collapse
Affiliation(s)
- Parisa Fayyazpour
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Fayyazpour
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yosra Vaez-Gharamaleki
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mortaza Raeisi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
37
|
Nguyen Cao TG, Truong Hoang Q, Kang JH, Kang SJ, Ravichandran V, Rhee WJ, Lee M, Ko YT, Shim MS. Bioreducible exosomes encapsulating glycolysis inhibitors potentiate mitochondria-targeted sonodynamic cancer therapy via cancer-targeted drug release and cellular energy depletion. Biomaterials 2023; 301:122242. [PMID: 37473534 DOI: 10.1016/j.biomaterials.2023.122242] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 07/22/2023]
Abstract
Nanocarrier-assisted sonodynamic therapy (SDT) has shown great potential for the effective and targeted treatment of deep-seated tumors by overcoming the critical limitations of sonosensitizers. However, in vivo SDT using nanocarriers is still constrained by their intrinsic toxicity and nonspecific cargo release. In this study, we developed bioreducible exosomes for the safe and tumor-specific delivery of mitochondria-targeting sonosensitizers [triphenylphosphonium-conjugated chlorin e6 (T-Ce6)] and glycolysis inhibitors (FX11). Redox-cleavable diselenide linker-bearing lipids were embedded into exosomes to trigger drug release in response to overexpressed glutathione in the tumor microenvironment. Bioreducible exosomes facilitate the cytoplasmic release of their payload in the reducing environment of tumor cells. They significantly enhance drug release and sonodynamic effects when irradiated with ultrasound (US). The mitochondria-targeted accumulation of T-Ce6 efficiently damaged the mitochondria of the cells under US irradiation, accelerating apoptotic cell death. FX11 substantially inhibited cellular energy metabolism, potentiating the antitumor efficacy of mitochondria-targeted SDT. Bioreducible exosomes effectively suppressed tumor growth in mice without significant systemic toxicity, via a combination of mitochondria-targeted SDT and energy metabolism-targeted therapy. This study offers new insights into the use of dual stimuli-responsive exosomes encapsulating sonosensitizers for safe and targeted sonodynamic cancer therapy.
Collapse
Affiliation(s)
- Thuy Giang Nguyen Cao
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Quan Truong Hoang
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Ji Hee Kang
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Su Jin Kang
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Vasanthan Ravichandran
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea; Research Center for Bio Materials & Process Development, Incheon National University, 119 Academy-ro, Yeonsu-gu, Incheon, 22012, Republic of Korea.
| | - Minjong Lee
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea; Department of Internal Medicine, Ewha Womans University Medical Center, Seoul, 07804, Republic of Korea.
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea.
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea.
| |
Collapse
|
38
|
Tashak Golroudbari H, Banikarimi SP, Ayati A, Hadizadeh A, Khorasani Zavareh Z, Hajikhani K, Heirani-Tabasi A, Ahmadi Tafti M, Davoodi S, Ahmadi Tafti H. Advanced micro-/nanotechnologies for exosome encapsulation and targeting in regenerative medicine. Clin Exp Med 2023; 23:1845-1866. [PMID: 36705868 DOI: 10.1007/s10238-023-00993-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/05/2023] [Indexed: 01/28/2023]
Abstract
Exosomes, a subset of vesicles generated from cell membranes, are crucial for cellular communication. Exosomes' innate qualities have been used in recent studies to create nanocarriers for various purposes, including medication delivery and immunotherapy. As a result, a wide range of approaches has been designed to utilize their non-immunogenic nature, drug-loading capacity, or targeting ability. In this study, we aimed to review the novel methods and approaches in exosome engineering for encapsulation and targeting in regenerative medicine. We have assessed and evaluated each method's efficacy, advantages, and disadvantages and discussed the results of related studies. Even though the therapeutic role of non-allogenic exosomes has been demonstrated in several studies, their application has certain limitations as these particles are neither fully specific to target tissue nor tissue retainable. Hence, there is a strong demand for developing more efficient encapsulation methods along with more accurate and precise targeting methods, such as 3D printing and magnetic nanoparticle loading in exosomes, respectively.
Collapse
Affiliation(s)
- Hasti Tashak Golroudbari
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyedeh Parnian Banikarimi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Regenerative Medicine, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Aryan Ayati
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Hadizadeh
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Khorasani Zavareh
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Kiana Hajikhani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Asieh Heirani-Tabasi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Ahmadi Tafti
- Colorectal Surgery Research Center, Imam Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Davoodi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ahmadi Tafti
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Zeng Y, Hu S, Luo Y, He K. Exosome Cargos as Biomarkers for Diagnosis and Prognosis of Hepatocellular Carcinoma. Pharmaceutics 2023; 15:2365. [PMID: 37765333 PMCID: PMC10537613 DOI: 10.3390/pharmaceutics15092365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Due to the insidiousness of HCC onset and the lack of specific early-stage markers, the early diagnosis and treatment of HCC are still unsatisfactory, leading to a poor prognosis. Exosomes are a type of extracellular vesicle containing various components, which play an essential part in the development, progression, and metastasis of HCC. A large number of studies have demonstrated that exosomes could serve as novel biomarkers for the diagnosis of HCC. These diagnostic components mainly include proteins, microRNAs, long noncoding RNAs, and circular RNAs. The exosome biomarkers showed high sensitivity and high specificity in distinguishing HCC from health controls and other liver diseases, such as chronic HBV and liver cirrhosis. The expression of these biomarkers also exhibits correlations with various clinical factors such as tumor size, TMN stage, overall survival, and recurrence rate. In this review, we summarize the function of exosomes in the development of HCC and highlight their application as HCC biomarkers for diagnosis and prognosis prediction.
Collapse
Affiliation(s)
- Yulai Zeng
- Department of Liver Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.Z.); (S.H.)
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
- Shanghai Institute of Transplantation, Shanghai 200127, China
| | - Shuyu Hu
- Department of Liver Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.Z.); (S.H.)
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
- Shanghai Institute of Transplantation, Shanghai 200127, China
| | - Yi Luo
- Department of Liver Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.Z.); (S.H.)
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
- Shanghai Institute of Transplantation, Shanghai 200127, China
| | - Kang He
- Department of Liver Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai 200127, China; (Y.Z.); (S.H.)
- Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200127, China
- Shanghai Institute of Transplantation, Shanghai 200127, China
| |
Collapse
|
40
|
Wu Q, Li L, Jia Y, Xu T, Zhou X. Advances in studies of circulating microRNAs: origination, transportation, and distal target regulation. J Cell Commun Signal 2023; 17:445-455. [PMID: 36357651 PMCID: PMC9648873 DOI: 10.1007/s12079-022-00705-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/07/2022] [Indexed: 11/12/2022] Open
Abstract
In the past few years, numerous advances emerged in terms of circulating microRNA(miRNA) regulating gene expression by circulating blood to the distal tissues and cells. This article reviewed and summarized the process of circulating miRNAs entering the circulating system to exert gene regulation, especially exogenous miRNAs (such as plant miRNAs), from the perspective of the circulating miRNAs source (cell secretion or gastrointestinal absorption), the transport form and pharmacokinetics in circulating blood, and the evidence of distal regulation to gene expression, thereby providing a basis for their in-depth research and even application prospects.
Collapse
Affiliation(s)
- Qingni Wu
- Evidence Based Medicine Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Longxue Li
- Laboratory Animal Science and Technology Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
- Key Laboratory of Animal Model of TCM Syndromes of Depression, Jiangxi Administration of traditional Chinese Medicine, 330004, Nanchang, China
| | - Yao Jia
- Evidence Based Medicine Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Tielong Xu
- Evidence Based Medicine Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China.
| | - Xu Zhou
- Evidence Based Medicine Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, 610000, Chengdu, China.
| |
Collapse
|
41
|
Ngu A, Munir J, Zempleni J. Milk-borne small extracellular vesicles: kinetics and mechanisms of transport, distribution, and elimination. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:339-346. [PMID: 37829291 PMCID: PMC10568984 DOI: 10.20517/evcna.2023.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Small extracellular vesicles (sEVs) in milk have the qualities desired for delivering therapeutics to diseased tissues. The production of bovine milk sEVs is scalable (1021 annually per cow), and they resist degradation in the gastrointestinal tract. Most cells studied to date internalize milk sEVs by a saturable process that follows Michaelis-Menten kinetics. The bioavailability of oral milk sEVs is approximately 50%. In addition to crossing the intestinal mucosa, milk sEVs also cross barriers such as the placenta and blood-brain barrier, thereby enabling the delivery of therapeutics to hard-to-reach tissues. In time course studies, levels of milk sEVs peaked in the intestinal mucosa, plasma, and urine approximately 6 h and returned to baseline 24 h after oral gavage in mice. In tissues, milk sEV levels peaked 12 h after gavage. Milk sEVs appear to be biologically safe. No cytokine storm was observed when milk sEVs were added to cultures of human peripheral blood mononuclear cells or administered orally to rats. Liver and kidney function and erythropoiesis were not impaired when milk sEVs were administered to rats by oral gavage for up to 15 days. Protocols for loading milk sEVs with therapeutic cargo are available. Currently, the use of milk sEVs (and other nanoparticles) in the delivery of therapeutics is limited by their rapid elimination through internalization by macrophages and lysosomal degradation in target cells. This mini review discusses the current knowledge base of sEV tissue distribution, excretion in feces and urine, internalization by macrophages, and degradation in lysosomes.
Collapse
Affiliation(s)
- Alice Ngu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0806, USA
| | - Javaria Munir
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0806, USA
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583-0806, USA
| |
Collapse
|
42
|
Yu W, Nan X, Schroyen M, Wang Y, Xiong B. Inulin-induced differences on serum extracellular vesicles derived miRNAs in dairy cows suffering from subclinical mastitis. Animal 2023; 17:100954. [PMID: 37690274 DOI: 10.1016/j.animal.2023.100954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
MicroRNA (miRNA) profiles vary with the nutritional and pathological conditions of cattle. In this study, we aimed to investigate the effects of inulin supplement on miRNA profiles derived from serum extracellular vesicles (EVs). Our goal was to determine the differences in miRNA expressions and analyse the pathways in which they are involved. Based on the results of California mastitis test and milk somatic cell counts, ten lactating cows with subclinical mastitis were randomly divided into two groups: an inulin group and a control group (n = 5 in each group). The inulin group received a daily supplement of 300 g of inulin while the control group did not receive any supplementation. After a 5-week treatment period, serum-derived EV-miRNAs from each cow were isolated. High-throughput sequencing was conducted to identify differentially expressed miRNAs. GO and KEGG bioinformatics analysis was performed to examine the target genes of these differentially expressed miRNAs. The EV-RNA concentration and small RNA content were not affected by the inulin treatment. A total of 162 known miRNAs and 180 novel miRNAs were identified from 10 samples in the two groups. Among the known miRNAs, 23 miRNAs were found to be differentially expressed between the two groups, with 18 upregulated and five downregulated in the inulin group compared to the control group. Pathway analysis revealed the involvement of these differentially expressed miRNAs in the regulation of cell structure and function, lipid oxidation and metabolism, immunity and inflammation, as well as digestion and absorption of nutrients. Overall, our study provides a molecular-level explanation for the reported beneficial health effects of inulin supplementation in cows with subclinical mastitis.
Collapse
Affiliation(s)
- W Yu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China; Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - X Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - M Schroyen
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Y Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - B Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| |
Collapse
|
43
|
Chancharoenthana W, Traitanon O, Leelahavanichkul A, Tasanarong A. Molecular immune monitoring in kidney transplant rejection: a state-of-the-art review. Front Immunol 2023; 14:1206929. [PMID: 37675106 PMCID: PMC10477600 DOI: 10.3389/fimmu.2023.1206929] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 07/31/2023] [Indexed: 09/08/2023] Open
Abstract
Although current regimens of immunosuppressive drugs are effective in renal transplant recipients, long-term renal allograft outcomes remain suboptimal. For many years, the diagnosis of renal allograft rejection and of several causes of renal allograft dysfunction, such as chronic subclinical inflammation and infection, was mostly based on renal allograft biopsy, which is not only invasive but also possibly performed too late for proper management. In addition, certain allograft dysfunctions are difficult to differentiate from renal histology due to their similar pathogenesis and immune responses. As such, non-invasive assays and biomarkers may be more beneficial than conventional renal biopsy for enhancing graft survival and optimizing immunosuppressive drug regimens during long-term care. This paper discusses recent biomarker candidates, including donor-derived cell-free DNA, transcriptomics, microRNAs, exosomes (or other extracellular vesicles), urine chemokines, and nucleosomes, that show high potential for clinical use in determining the prognosis of long-term outcomes of kidney transplantation, along with their limitations.
Collapse
Affiliation(s)
- Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Tropical Immunology and Translational Research Unit (TITRU), Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Thammasat Multi-Organ Transplant Center, Thammasat University Hospital, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Opas Traitanon
- Thammasat Multi-Organ Transplant Center, Thammasat University Hospital, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Adis Tasanarong
- Thammasat Multi-Organ Transplant Center, Thammasat University Hospital, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
44
|
Lau SY, Kang M, Hisey CL, Chamley LW. Studying exogenous extracellular vesicle biodistribution by in vivo fluorescence microscopy. Dis Model Mech 2023; 16:dmm050074. [PMID: 37526034 PMCID: PMC10417515 DOI: 10.1242/dmm.050074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bound vesicles released from cells that play a crucial role in many physiological processes and pathological mechanisms. As such, there is great interest in their biodistribution. One currently accessible technology to study their fate in vivo involves fluorescent labelling of exogenous EVs followed by whole-animal imaging. Although this is not a new technology, its translation from studying the fate of whole cells to subcellular EVs requires adaptation of the labelling techniques, excess dye removal and a refined experimental design. In this Review, we detail the methods and considerations for using fluorescence in vivo and ex vivo imaging to study the biodistribution of exogenous EVs and their roles in physiology and disease biology.
Collapse
Affiliation(s)
- Sien Yee Lau
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
| | - Matthew Kang
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
| | - Colin L. Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Lawrence W. Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland 1023, New Zealand
- Hub for Extracellular Vesicle Investigations, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
45
|
Banerjee A, Lino M, Jesus C, Ribeiro Q, Abrunhosa A, Ferreira L. Imaging platforms to dissect the in vivo communication, biodistribution and controlled release of extracellular vesicles. J Control Release 2023; 360:549-563. [PMID: 37406818 DOI: 10.1016/j.jconrel.2023.06.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Extracellular vesicles (EVs) work as communication vehicles, allowing the exchange of bioactive molecules (microRNAs, mRNAs, proteins, etc) between neighbouring and distant cells in the organism. EVs are thus important players in several physiological and pathological processes. Thus, it is critical to understand their role in cellular/organ communication to fully evaluate their biological, diagnosis and therapeutic potential. In addition, recent studies have explored the controlled release of EVs for regenerative medicine applications and thus the evaluation of their release profile is important to correlate with biological activity. Here, we give a brief introduction about EV imaging platforms in terms of their sensitivity, penetration depth, cost, and operational simplicity, followed by a discussion of different EV labelling processes with their advantages and limitations. Next, we cover the relevance of these imaging platforms to dissect the tropism and biological role of endogenous EVs. We also cover the relevance of imaging platforms to monitor the accumulation of exogenous EVs and their potential cellular targets. Finally, we highlight the importance of imaging platforms to investigate the release profile of EVs from different controlled systems.
Collapse
Affiliation(s)
- Arnab Banerjee
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Miguel Lino
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Carlos Jesus
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Quélia Ribeiro
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Antero Abrunhosa
- ICNAS/CIBIT - Institute for Nuclear Sciences Applied to Health/Coimbra Institute for Biomedical Imaging and Translational research, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Lino Ferreira
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| |
Collapse
|
46
|
Lin SW, Tsai JC, Shyong YJ. Drug delivery of extracellular vesicles: Preparation, delivery strategies and applications. Int J Pharm 2023; 642:123185. [PMID: 37391106 DOI: 10.1016/j.ijpharm.2023.123185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Extracellular vesicles (EV) are cell-originated vesicles exhibited with characteristics similar to the parent cells. Several studies have suggested the therapeutic potential of EV since they played as an intercellular communicator and modulate disease microenvironment, and thus EV has been widely studied in cancer management and tissue regeneration. However, merely application of EV revealed limited therapeutic outcome in different disease scenario and co-administration of drugs may be necessary to exert proper therapeutic effect. The method of drug loading into EV and efficient delivery of the formulation is therefore important. In this review, the advantages of using EV as drug delivery system compared to traditional synthetic nanoparticles will be emphasized, followed by the method of preparing EV and drug loading. The pharmacokinetic characteristics of EV was discussed, together with the review of reported delivery strategies and related application of EV in different disease management.
Collapse
Affiliation(s)
- Shang-Wen Lin
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| |
Collapse
|
47
|
Cieślik M, Bryniarski K, Nazimek K. Biodelivery of therapeutic extracellular vesicles: should mononuclear phagocytes always be feared? Front Cell Dev Biol 2023; 11:1211833. [PMID: 37476156 PMCID: PMC10354279 DOI: 10.3389/fcell.2023.1211833] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023] Open
Abstract
At present, extracellular vesicles (EVs) are considered key candidates for cell-free therapies, including treatment of allergic and autoimmune diseases. However, their therapeutic effectiveness, dependent on proper targeting to the desired cells, is significantly limited due to the reduced bioavailability resulting from their rapid clearance by the cells of the mononuclear phagocyte system (MPS). Thus, developing strategies to avoid EV elimination is essential when applying them in clinical practice. On the other hand, malfunctioning MPS contributes to various immune-related pathologies. Therapeutic reversal of these effects with EVs would be beneficial and could be achieved, for example, by modulating the macrophage phenotype or regulating antigen presentation by dendritic cells. Additionally, intended targeting of EVs to MPS macrophages for replication and repackaging of their molecules into new vesicle subtype can allow for their specific targeting to appropriate populations of acceptor cells. Herein, we briefly discuss the under-explored aspects of the MPS-EV interactions that undoubtedly require further research in order to accelerate the therapeutic use of EVs.
Collapse
Affiliation(s)
| | | | - Katarzyna Nazimek
- Department of Immunology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
48
|
Li X, Wu J, Wu Y, Duan Z, Luo M, Li L, Li S, Jia Y. Imbalance of Vaginal Microbiota and Immunity: Two Main Accomplices of Cervical Cancer in Chinese Women. Int J Womens Health 2023; 15:987-1002. [PMID: 37424699 PMCID: PMC10329453 DOI: 10.2147/ijwh.s406596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Objective To explore the correlation of female vaginal microbiota and immune factors with cervical cancer. Methods The distribution pattern difference of vaginal microbiota of four groups of women (cervical cancer, HPV-positive CIN, HPV-positive non-CIN, and HPV-negative groups) were compared by microbial 16S rDNA sequencing. The protein chip was used to detect the composition and changes of the immune factors in the four groups. Results Alpha diversity analysis demonstrated that the diversity of the vaginal microbiota was increased as the disease develops. Among those bacteria abundant in the vaginal microbiota, Lactobacillus, Prevotella, and Gardnerella dominate at the genus level of vaginal flora. Compared with the HPV-negative group, the differentially dominant bacteria, such as Prevotella, Ralstonia, Gardnerella and Sneathia, are enriched in the cervical cancer group. Likewise, Gardnerella, Prevotella, and Sneathia are more in the HPV-positive CIN group, while Gardnerella and Prevotella in the HPV-positive non-CIN group, respectively. In contrast, Lactobacillus and Atopobium are dominant in the HPV-negative group (LDA>4log10). The concentration of inflammatory immune factors IP-10 and VEGF-A were increased in the cervical cancer group (P < 0.05), compared with other groups. Conclusion The occurrence of cervical cancer is related to an increase of vaginal microbiota diversity and up-regulation of inflammatory immune factor proteins. The abundance of Lactobacillus was decreased while the one of Prevotella and Gardnerella were increased in the cervical cancer group, compared with other three groups. Moreover, the IP-10 and VEGF-A were also increased in the cervical cancer group. Thus, evaluation of changes in the vaginal microbiota and these two immune factor levels might be a potential non-invasive and simple method to predict cervical cancer. Furthermore, it is significant to adjust and restore the balance of vaginal microbiota and maintain normal immune function in preventing and treating cervical cancer.
Collapse
Affiliation(s)
- Xiaoge Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Jin Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Yutong Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Zhaoning Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Ming Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Ling Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Sijing Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| | - Ying Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People’s Republic of China
| |
Collapse
|
49
|
Wijenayake S, Martz J, Lapp HE, Storm JA, Champagne FA, Kentner AC. The contributions of parental lactation on offspring development: It's not udder nonsense! Horm Behav 2023; 153:105375. [PMID: 37269591 PMCID: PMC10351876 DOI: 10.1016/j.yhbeh.2023.105375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/05/2023]
Abstract
The Developmental Origins of Health and Disease (DOHaD) hypothesis describes how maternal stress exposures experienced during critical periods of perinatal life are linked to altered developmental trajectories in offspring. Perinatal stress also induces changes in lactogenesis, milk volume, maternal care, and the nutritive and non-nutritive components of milk, affecting short and long-term developmental outcomes in offspring. For instance, selective early life stressors shape the contents of milk, including macro/micronutrients, immune components, microbiota, enzymes, hormones, milk-derived extracellular vesicles, and milk microRNAs. In this review, we highlight the contributions of parental lactation to offspring development by examining changes in the composition of breast milk in response to three well-characterized maternal stressors: nutritive stress, immune stress, and psychological stress. We discuss recent findings in human, animal, and in vitro models, their clinical relevance, study limitations, and potential therapeutic significance to improving human health and infant survival. We also discuss the benefits of enrichment methods and support tools that can be used to improve milk quality and volume as well as related developmental outcomes in offspring. Lastly, we use evidence-based primary literature to convey that even though select maternal stressors may modulate lactation biology (by influencing milk composition) depending on the severity and length of exposure, exclusive and/or prolonged milk feeding may attenuate the negative in utero effects of early life stressors and promote healthy developmental trajectories. Overall, scientific evidence supports lactation to be protective against nutritive and immune stressors, but the benefits of lactation in response to psychological stressors need further investigation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
| | - Julia Martz
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Hannah E Lapp
- Deparment of Psychology, University of Texas at Austin, Austin, TX, USA
| | - Jasmyne A Storm
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada
| | | | - Amanda C Kentner
- School of Arts & Sciences, Health Psychology Program, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA.
| |
Collapse
|
50
|
Moholkar DN, Kandimalla R, Gupta RC, Aqil F. Advances in lipid-based carriers for cancer therapeutics: Liposomes, exosomes and hybrid exosomes. Cancer Lett 2023; 565:216220. [PMID: 37209944 PMCID: PMC10325927 DOI: 10.1016/j.canlet.2023.216220] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/18/2023] [Accepted: 05/06/2023] [Indexed: 05/22/2023]
Abstract
Cancer has recently surpassed heart disease as the leading cause of deaths worldwide for the age group 45-65 and has been the primary focus for biomedical researchers. Presently, the drugs involved in the first-line cancer therapy are raising concerns due to high toxicity and lack of selectivity to cancer cells. There has been a significant increase in research with innovative nano formulations to entrap the therapeutic payload to enhance efficacy and eliminate or minimize toxic effects. Lipid-based carriers stand out due to their unique structural properties and biocompatible nature. The two main leaders of lipid-based drug carriers: long known liposomes and comparatively new exosomes have been well-researched. The similarity between the two lipid-based carriers is the vesicular structure with the core's capability to carry the payload. While liposomes utilize chemically derived and altered phospholipid components, the exosomes are naturally occurring vesicles with inherent lipids, proteins, and nucleic acids. More recently, researchers have focused on developing hybrid exosomes by fusing liposomes and exosomes. Combining these two types of vesicles may offer some advantages such as high drug loading, targeted cellular uptake, biocompatibility, controlled release, stability in harsh conditions and low immunogenicity.
Collapse
Affiliation(s)
- Disha N Moholkar
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Raghuram Kandimalla
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Ramesh C Gupta
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA; Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| | - Farrukh Aqil
- Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA; Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|