1
|
Murphy CA, O'Reilly D, Weiss L, Madden S, Macleod H, Chevillier AL, Neary E, O'Loughlin J, EL‐Khuffash A, Kevane B, NíAinle F, Zivny J, McCallion N, Maguire PB. Unique Patterns of Circulating Extracellular Vesicles in Preterm Infants During Adaptation to Extra-Uterine Life. J Extracell Vesicles 2025; 14:e70064. [PMID: 40194994 PMCID: PMC11975508 DOI: 10.1002/jev2.70064] [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: 07/25/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
There is growing interest in the role of extracellular vesicles (EVs) in neonatal pathology. This study aimed to characterise circulating EVs following preterm birth. This single-centre prospective observational study included cord and postnatal plasma from preterm (n = 101) and full-term infants (n = 66). EVs were analysed using nanoparticle tracking analysis, flow cytometry, proteomics and procoagulant activity assay. We found changes in the concentration, size, cellular origin and proteomic content of circulating EVs in preterm infants during perinatal adaptation. To understand if these changes were related to prematurity or normal adaptation to extrauterine life, they were also investigated in term infants. There was a dramatic increase in the concentration of small and large EVs on Day 3 in the preterm group; specific subsets of platelet (CD42b+ and CD62P+), endothelial (VEGFR2) and tissue factor EVs were elevated. Differentially expressed proteins relating to haemostasis, pulmonary physiology and immunity were identified between Day 1 and 3 in preterm infants. These changes have never previously been described in a large cohort of preterm infants and differ from healthy term infants. These findings have major implications for future neonatal EV studies, particularly the timing of sample collection. Further work is required to understand the clinical implications of this unique EV profile following preterm birth.
Collapse
Affiliation(s)
- Claire A. Murphy
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Daniel O'Reilly
- Department of NeonatologyRotunda HospitalDublinIreland
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Luisa Weiss
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Stephen Madden
- School of Population HealthRoyal College of Surgeons in IrelandDublinIreland
| | - Hayley Macleod
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Ana Le Chevillier
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| | - Elaine Neary
- Department of NeonatologyLiverpool Women's HospitalLiverpoolUK
- Department of Health and Life SciencesUniversity of LiverpoolLiverpoolUK
| | - John O'Loughlin
- Department of Laboratory MedicineRotunda HospitalDublinIreland
| | - Afif EL‐Khuffash
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Barry Kevane
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| | - Fionnuala NíAinle
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
- Department of HaematologyRotunda HospitalDublinIreland
| | - Jan Zivny
- Institute of Pathological PhysiologyFirst Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Naomi McCallion
- Department of PaediatricsRoyal College of Surgeons in IrelandDublinIreland
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Patricia B. Maguire
- Conway‐SPHERE Research Group, Conway InstituteUniversity College DublinDublinIreland
| |
Collapse
|
2
|
Staub E, Cao Q, Chen XM, Pollock C. Concentration of kidney markers and detection of exosomes in urine samples collected in cotton wool balls in preterm and term neonates. Pathology 2025; 57:81-86. [PMID: 39516170 DOI: 10.1016/j.pathol.2024.07.006] [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: 03/15/2024] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 11/16/2024]
Abstract
Collecting urine samples in neonates by catheterisation or suprapubic puncture causes trauma, whereas self-adhesive collection bags can damage fragile skin. An alternative method is the collection of samples from urine-soaked cotton wool balls placed in diapers. The aim of this study was to compare the concentration of albumin, creatinine, neutrophil gelatinase-associated lipocalin (NGAL), and uromodulin between clean-catch urine and samples collected in cotton wool balls in neonates and assess the efficiency of exosome extraction. Standard clean-catch urine samples were assayed for albumin, creatinine, NGAL, and uromodulin using commercial enzyme-linked immunosorbent assay (ELISA) kits. Concentrations were compared to the same urine samples extracted immediately from soaked cotton wool balls (sample 2, S2) or the urine extracted from cotton wool balls placed in a diaper in a warm incubator for 2 h before extraction (sample 3, S3). Exosomes were extracted from all three samples of one patient for visualisation by electron microscopy. Twenty-six infants (17 males) of median gestational age at birth of 32+1 weeks had urine collected at a median age of 29 days at 37+6 weeks corrected age. Concentrations in S2 and S3 were within 10% of the concentration of standard samples in 46% and 35% of specimens for albumin, 69% and 58% for creatinine, 12% and 12% for NGAL, and 27% and 15% for uromodulin, respectively, without consistent positive or negative bias. Urine albumin/creatinine ratios (UACRs) were 4.3% less in S2 and 4.5% less in S3 than in standard samples. Exosomes were extracted and visualised from all three sample types. Neonatal urine samples extracted from cotton wool balls can be used to screen for relevant albuminuria but provide imprecise estimates of NGAL and uromodulin. The proof of exosome extraction from urine collection in cotton wool balls opens the potential to examine exosomal cargo.
Collapse
Affiliation(s)
- Eveline Staub
- Department of Neonatology, Royal North Shore Hospital, St Leonards, NSW, Australia; Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, Australia.
| | - Qinghua Cao
- Renal Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Xin-Ming Chen
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, Australia; Renal Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Carol Pollock
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW, Australia; Renal Research, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
3
|
Shanmugam I, Radhakrishnan S, Santosh S, Ramnath A, Anil M, Devarajan Y, Maheswaran S, Narayanan V, Pitchaimani A. Emerging role and translational potential of small extracellular vesicles in neuroscience. Life Sci 2024; 355:122987. [PMID: 39151884 DOI: 10.1016/j.lfs.2024.122987] [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: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Small extracellular vesicles (sEV) are endogenous lipid-bound membrane vesicles secreted by both prokaryotic and eukaryotic cells into the extracellular environment, performs several biological functions such as cell-cell communication, transfer of proteins, mRNA, and ncRNA to target cells in distant sites. Due to their role in molecular pathogenesis and its potential to deliver biological cargo to target cells, it has become a prominent area of interest in recent research in the field of Neuroscience. However, their role in neurological disorders, like neurodegenerative diseases is more complex and still unaddressed. Thus, this review focuses on the role of sEV in neurodegenerative and neurodevelopmental diseases, including their biogenesis, classification, and pathogenesis, with translational advantages and limitations in the area of neurobiology.
Collapse
Affiliation(s)
- Iswarya Shanmugam
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sivani Radhakrishnan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Shradha Santosh
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Akansha Ramnath
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Meghna Anil
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Yogesh Devarajan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Saravanakumar Maheswaran
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Vaibav Narayanan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
4
|
Kumar N, Bidkhori HR, Yawno T, Lim R, Inocencio IM. Therapeutic potential of extracellular vesicles derived from human amniotic epithelial cells for perinatal cerebral and pulmonary injury. Stem Cells Transl Med 2024; 13:711-723. [PMID: 38895873 PMCID: PMC11328935 DOI: 10.1093/stcltm/szae037] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/21/2024] Open
Abstract
Lung and brain injury that occurs during the perinatal period leads to lifelong disability and is often driven and/or exacerbated by inflammation. Human amniotic epithelial cells (hAEC), which demonstrate immunomodulatory, anti-fibrotic, and regenerative capabilities, are being explored as a therapeutic candidate for perinatal injury. However, limitations regarding scalable manufacturing, storage, transport, and dose-related toxicity have impeded clinical translation. Isolated therapeutic extracellular vesicles (EVs) from stem and stem-like cells are thought to be key paracrine mediators of therapeutic efficacy. The unique characteristics of EVs suggest that they potentially circumvent the limitations of traditional cell-based therapies. However, given the novelty of EVs as a therapeutic, recommendations around ideal methods of production, isolation, storage, and delivery have not yet been created by regulatory agencies. In this concise review, we discuss the pertinence and limitations of cell-based therapeutics in perinatal medicine. We also review the preclinical evidence supporting the use of therapeutic EVs for perinatal therapy. Further, we summarize the arising considerations regarding adequate cell source, biodistribution, isolation and storage methods, and regulatory roadblocks for the development of therapeutic EVs.
Collapse
Affiliation(s)
- Naveen Kumar
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Hamid Reza Bidkhori
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Tamara Yawno
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
- Department of Paediatrics, Monash University, Clayton 3168, Victoria, Australia
| | - Rebecca Lim
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| | - Ishmael Miguel Inocencio
- The Ritchie Centre, The Hudson Institute of Medical Research, Clayton 3168, Victoria, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton 3168, Victoria, Australia
| |
Collapse
|
5
|
Antounians L, Figueira RL, Kukreja B, Litvack ML, Zani-Ruttenstock E, Khalaj K, Montalva L, Doktor F, Obed M, Blundell M, Wu T, Chan C, Wagner R, Lacher M, Wilson MD, Post M, Kalish BT, Zani A. Fetal hypoplastic lungs have multilineage inflammation that is reversed by amniotic fluid stem cell extracellular vesicle treatment. SCIENCE ADVANCES 2024; 10:eadn5405. [PMID: 39058789 PMCID: PMC11277482 DOI: 10.1126/sciadv.adn5405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/21/2024] [Indexed: 07/28/2024]
Abstract
Antenatal administration of extracellular vesicles from amniotic fluid stem cells (AFSC-EVs) reverses features of pulmonary hypoplasia in models of congenital diaphragmatic hernia (CDH). However, it remains unknown which lung cellular compartments and biological pathways are affected by AFSC-EV therapy. Herein, we conducted single-nucleus RNA sequencing (snRNA-seq) on rat fetal CDH lungs treated with vehicle or AFSC-EVs. We identified that intra-amniotically injected AFSC-EVs reach the fetal lung in rats with CDH, where they promote lung branching morphogenesis and epithelial cell differentiation. Moreover, snRNA-seq revealed that rat fetal CDH lungs have a multilineage inflammatory signature with macrophage enrichment, which is reversed by AFSC-EV treatment. Macrophage enrichment in CDH fetal rat lungs was confirmed by immunofluorescence, flow cytometry, and inhibition studies with GW2580. Moreover, we validated macrophage enrichment in human fetal CDH lung autopsy samples. Together, this study advances knowledge on the pathogenesis of pulmonary hypoplasia and further evidence on the value of an EV-based therapy for CDH fetuses.
Collapse
Affiliation(s)
- Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Bharti Kukreja
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Michael L. Litvack
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Elke Zani-Ruttenstock
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Louise Montalva
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Fabian Doktor
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Mikal Obed
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Matisse Blundell
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Taiyi Wu
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
| | - Cadia Chan
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Richard Wagner
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Martin Lacher
- Department of Pediatric Surgery, Leipzig University, Leipzig 04109, Germany
| | - Michael D. Wilson
- Genetics and Genome Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
| | - Martin Post
- Translational Medicine Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5T 1P5, Canada
| | - Brian T. Kalish
- Neurosciences and Mental Health Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto M5S 1A8, Canada
- Division of Neonatology, The Hospital for Sick Children, Toronto M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto M5G 1X8, Canada
- Department of Surgery, University of Toronto, Toronto M5T 1P5, Canada
| |
Collapse
|
6
|
Biouss G, Antounians L, Aguet J, Kopcalic K, Fakhari N, Baranger J, Mertens L, Villemain O, Zani A. The brain of fetuses with congenital diaphragmatic hernia shows signs of hypoxic injury with loss of progenitor cells, neurons, and oligodendrocytes. Sci Rep 2024; 14:13680. [PMID: 38871804 DOI: 10.1038/s41598-024-64412-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 06/08/2024] [Indexed: 06/15/2024] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a birth defect characterized by incomplete closure of the diaphragm, herniation of abdominal organs into the chest, and compression of the lungs and the heart. Besides complications related to pulmonary hypoplasia, 1 in 4 survivors develop neurodevelopmental impairment, whose etiology remains unclear. Using a fetal rat model of CDH, we demonstrated that the compression exerted by herniated organs on the mediastinal structures results in decreased brain perfusion on ultrafast ultrasound, cerebral hypoxia with compensatory angiogenesis, mature neuron and oligodendrocyte loss, and activated microglia. In CDH fetuses, apoptosis was prominent in the subventricular and subgranular zones, areas that are key for neurogenesis. We validated these findings in the autopsy samples of four human fetuses with CDH compared to age- and sex-matched controls. This study reveals the molecular mechanisms and cellular changes that occur in the brain of fetuses with CDH and creates opportunities for therapeutic targets.
Collapse
Affiliation(s)
- George Biouss
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Julien Aguet
- Department of Diagnostic Imaging, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Medical Imaging, University of Toronto, 263 McCaul Street, Toronto, ON, M5T 1W7, Canada
| | - Katarina Kopcalic
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Nikan Fakhari
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Jerome Baranger
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Luc Mertens
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Olivier Villemain
- Translation Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Pediatrics, Labatt Family Heart Centre, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Division of General and Thoracic Surgery, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Surgery, University of Toronto, 200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
7
|
Fallahi S, Zangbar HS, Farajdokht F, Rahbarghazi R, Mohaddes G, Ghiasi F. Exosomes as a therapeutic tool to promote neurorestoration and cognitive function in neurological conditions: Achieve two ends with a single effort. CNS Neurosci Ther 2024; 30:e14752. [PMID: 38775149 PMCID: PMC11110007 DOI: 10.1111/cns.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/16/2024] [Accepted: 04/13/2024] [Indexed: 05/25/2024] Open
Abstract
Exosomes possess a significant role in intercellular communications. In the nervous system, various neural cells release exosomes that not only own a role in intercellular communications but also eliminate the waste of cells, maintain the myelin sheath, facilitate neurogenesis, and specifically assist in normal cognitive function. In neurological conditions including Parkinson's disease (PD), Alzheimer's disease (AD), traumatic brain injury (TBI), and stroke, exosomal cargo like miRNAs take part in the sequela of conditions and serve as a diagnostic tool of neurological disorders, too. Exosomes are not only a diagnostic tool but also their inhibition or administration from various sources like mesenchymal stem cells and serum, which have shown a worthy potential to treat multiple neurological disorders. In addition to neurodegenerative manifestations, cognitive deficiencies are an integral part of neurological diseases, and applying exosomes in improving both aspects of these diseases has been promising. This review discusses the status of exosome therapy in improving neurorestorative and cognitive function following neurological disease.
Collapse
Affiliation(s)
- Solmaz Fallahi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| | - Hamid Soltani Zangbar
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Fereshteh Farajdokht
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
| | - Gisou Mohaddes
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
- Department of Neuroscience and Cognition, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Neurosciences Research CenterTabriz University of Medical SciencesTabrizIran
- Department of Biomedical EducationCalifornia Health Sciences University, College of Osteopathic MedicineClovisCaliforniaUSA
| | - Fariba Ghiasi
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
- Department of PhysiologyTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
8
|
Goryunov K, Ivanov M, Kulikov A, Shevtsova Y, Burov A, Podurovskaya Y, Zubkov V, Degtyarev D, Sukhikh G, Silachev D. A Review of the Use of Extracellular Vesicles in the Treatment of Neonatal Diseases: Current State and Problems with Translation to the Clinic. Int J Mol Sci 2024; 25:2879. [PMID: 38474125 PMCID: PMC10932115 DOI: 10.3390/ijms25052879] [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: 01/15/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Neonatal disorders, particularly those resulting from prematurity, pose a major challenge in health care and have a significant impact on infant mortality and long-term child health. The limitations of current therapeutic strategies emphasize the need for innovative treatments. New cell-free technologies utilizing extracellular vesicles (EVs) offer a compelling opportunity for neonatal therapy by harnessing the inherent regenerative capabilities of EVs. These nanoscale particles, secreted by a variety of organisms including animals, bacteria, fungi and plants, contain a repertoire of bioactive molecules with therapeutic potential. This review aims to provide a comprehensive assessment of the therapeutic effects of EVs and mechanistic insights into EVs from stem cells, biological fluids and non-animal sources, with a focus on common neonatal conditions such as hypoxic-ischemic encephalopathy, respiratory distress syndrome, bronchopulmonary dysplasia and necrotizing enterocolitis. This review summarizes evidence for the therapeutic potential of EVs, analyzes evidence of their mechanisms of action and discusses the challenges associated with the implementation of EV-based therapies in neonatal clinical practice.
Collapse
Affiliation(s)
- Kirill Goryunov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Mikhail Ivanov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Andrey Kulikov
- Medical Institute, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Moscow 117198, Russia;
| | - Yulia Shevtsova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Artem Burov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Yulia Podurovskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Victor Zubkov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Dmitry Degtyarev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Denis Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|
9
|
Tscherrig V, Cottagnoud S, Haesler V, Renz P, Surbek D, Schoeberlein A, Joerger-Messerli MS. MicroRNA Cargo in Wharton's Jelly MSC Small Extracellular Vesicles: Key Functionality to In Vitro Prevention and Treatment of Premature White Matter Injury. Stem Cell Rev Rep 2023; 19:2447-2464. [PMID: 37523115 PMCID: PMC10579138 DOI: 10.1007/s12015-023-10595-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Preterm birth is the leading cause of childhood morbidity and mortality and can result in white matter injury (WMI), leading to long-term neurological disabilities with global health burden. Mesenchymal stromal cell-derived small extracellular vesicles (MSC-sEV) are a promising therapeutic agent for treating perinatal neurological injury. They carry microRNAs (miRNAs) predicted to be involved in the onset of premature WMI. We hypothesize that miRNAs have a key function in the beneficial effects of MSC-sEV. We isolated MSC from umbilical cord tissue, the Wharton's jelly (WJ), and purified small extracellular vesicles (sEV) from WJ-MSC culture supernatant by ultracentrifugation and size exclusion chromatography. The miRNA content was quantified by real-time polymerase chain reaction. A luciferase gene assay validated silencing of TP53 and TAOK1, which we previously identified as predicted target genes of MSC-sEV miRNAs by Next Generation Sequencing and pathway enrichment analysis. The impact of sEV miRNAs on oligodendroglial maturation and neuronal apoptosis was evaluated using an in vitro oxygen-glucose deprivation model (OGD/R) by knocking-down DROSHA in WJ-MSC, which initiates miRNA processing. WJ-MSC-sEV contained miRNAs involved in WMI, namely hsa-miR-22-3p, hsa-miR-21-5p, hsa-miR-27b-3p, and the hsa-let-7 family. The luciferase assay strongly indicated an inhibitory effect of sEV miRNAs on the gene expression of TP53 and TAOK1. Small EV initiated oligodendrocyte maturation and reduced OGD/R-mediated neuronal apoptosis. Knocking-down DROSHA in WJ-MSC reduced the expression of sEV miRNAs and led to the loss of their beneficial effects. Our in vitro study strongly indicates the key function of miRNAs in the therapeutic potential of WJ-MSC-sEV in premature WMI.
Collapse
Affiliation(s)
- Vera Tscherrig
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sophie Cottagnoud
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Valérie Haesler
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Patricia Renz
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Daniel Surbek
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Marianne Simone Joerger-Messerli
- Department of Obstetrics and Feto-maternal Medicine, University Women's Hospital, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
10
|
Wei X, Liu S, Cao Y, Wang Z, Chen S. Polymers in Engineering Extracellular Vesicle Mimetics: Current Status and Prospective. Pharmaceutics 2023; 15:pharmaceutics15051496. [PMID: 37242738 DOI: 10.3390/pharmaceutics15051496] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The maintenance of a high delivery efficiency by traditional nanomedicines during cancer treatment is a challenging task. As a natural mediator for short-distance intercellular communication, extracellular vesicles (EVs) have garnered significant attention owing to their low immunogenicity and high targeting ability. They can load a variety of major drugs, thus offering immense potential. In order to overcome the limitations of EVs and establish them as an ideal drug delivery system, polymer-engineered extracellular vesicle mimics (EVMs) have been developed and applied in cancer therapy. In this review, we discuss the current status of polymer-based extracellular vesicle mimics in drug delivery, and analyze their structural and functional properties based on the design of an ideal drug carrier. We anticipate that this review will facilitate a deeper understanding of the extracellular vesicular mimetic drug delivery system, and stimulate the progress and advancement of this field.
Collapse
Affiliation(s)
- Xinyue Wei
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Sihang Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Advanced Optical Communication Systems and Networks, Key Laboratory for Thin Film and Microfabrication of the Ministry of Education, UM-SJTU Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yifeng Cao
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Department of Electronic Chemicals, Institute of Zhejiang University-Quzhou, Quzhou 324000, China
| | - Zhen Wang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
- Zhejiang Sundoc Pharmaceutical Science and Tech Co., Ltd., Hangzhou 310051, China
| | - Shengfu Chen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
11
|
Spatio-temporal dynamics enhance cellular diversity, neuronal function and further maturation of human cerebral organoids. Commun Biol 2023; 6:173. [PMID: 36788328 PMCID: PMC9926461 DOI: 10.1038/s42003-023-04547-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
The bioengineerined and whole matured human brain organoids stand as highly valuable three-dimensional in vitro brain-mimetic models to recapitulate in vivo brain development, neurodevelopmental and neurodegenerative diseases. Various instructive signals affecting multiple biological processes including morphogenesis, developmental stages, cell fate transitions, cell migration, stem cell function and immune responses have been employed for generation of physiologically functional cerebral organoids. However, the current approaches for maturation require improvement for highly harvestable and functional cerebral organoids with reduced batch-to-batch variabilities. Here, we demonstrate two different engineering approaches, the rotating cell culture system (RCCS) microgravity bioreactor and a newly designed microfluidic platform (µ-platform) to improve harvestability, reproducibility and the survival of high-quality cerebral organoids and compare with those of traditional spinner and shaker systems. RCCS and µ-platform organoids have reached ideal sizes, approximately 95% harvestability, prolonged culture time with Ki-67 + /CD31 + /β-catenin+ proliferative, adhesive and endothelial-like cells and exhibited enriched cellular diversity (abundant neural/glial/ endothelial cell population), structural brain morphogenesis, further functional neuronal identities (glutamate secreting glutamatergic, GABAergic and hippocampal neurons) and synaptogenesis (presynaptic-postsynaptic interaction) during whole human brain development. Both organoids expressed CD11b + /IBA1 + microglia and MBP + /OLIG2 + oligodendrocytes at high levels as of day 60. RCCS and µ-platform organoids showing high levels of physiological fidelity a high level of physiological fidelity can serve as functional preclinical models to test new therapeutic regimens for neurological diseases and benefit from multiplexing.
Collapse
|
12
|
Schiller EA, Cohen K, Lin X, El-Khawam R, Hanna N. Extracellular Vesicle-microRNAs as Diagnostic Biomarkers in Preterm Neonates. Int J Mol Sci 2023; 24:2622. [PMID: 36768944 PMCID: PMC9916767 DOI: 10.3390/ijms24032622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Neonates born prematurely (<37 weeks of gestation) are at a significantly increased risk of developing inflammatory conditions associated with high mortality rates, including necrotizing enterocolitis, bronchopulmonary dysplasia, and hypoxic-ischemic brain damage. Recently, research has focused on characterizing the content of extracellular vesicles (EVs), particularly microRNAs (miRNAs), for diagnostic use. Here, we describe the most recent work on EVs-miRNAs biomarkers discovery for conditions that commonly affect premature neonates.
Collapse
Affiliation(s)
- Emily A. Schiller
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Koral Cohen
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Xinhua Lin
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
| | - Rania El-Khawam
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| | - Nazeeh Hanna
- Department of Foundational Medicine, New York University Long Island School of Medicine, Mineola, NY 11501, USA
- Department of Pediatrics, Division of Neonatology, New York University Langone Long Island Hospital, Mineola, NY 11501, USA
| |
Collapse
|
13
|
Al-Jipouri A, Almurisi SH, Al-Japairai K, Bakar LM, Doolaanea AA. Liposomes or Extracellular Vesicles: A Comprehensive Comparison of Both Lipid Bilayer Vesicles for Pulmonary Drug Delivery. Polymers (Basel) 2023; 15:318. [PMID: 36679199 PMCID: PMC9866119 DOI: 10.3390/polym15020318] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/31/2022] [Accepted: 01/01/2023] [Indexed: 01/11/2023] Open
Abstract
The rapid and non-invasive pulmonary drug delivery (PDD) has attracted great attention compared to the other routes. However, nanoparticle platforms, like liposomes (LPs) and extracellular vesicles (EVs), require extensive reformulation to suit the requirements of PDD. LPs are artificial vesicles composed of lipid bilayers capable of encapsulating hydrophilic and hydrophobic substances, whereas EVs are natural vesicles secreted by cells. Additionally, novel LPs-EVs hybrid vesicles may confer the best of both. The preparation methods of EVs are distinguished from LPs since they rely mainly on extraction and purification, whereas the LPs are synthesized from their basic ingredients. Similarly, drug loading methods into/onto EVs are distinguished whereby they are cell- or non-cell-based, whereas LPs are loaded via passive or active approaches. This review discusses the progress in LPs and EVs as well as hybrid vesicles with a special focus on PDD. It also provides a perspective comparison between LPs and EVs from various aspects (composition, preparation/extraction, drug loading, and large-scale manufacturing) as well as the future prospects for inhaled therapeutics. In addition, it discusses the challenges that may be encountered in scaling up the production and presents our view regarding the clinical translation of the laboratory findings into commercial products.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany
| | - Samah Hamed Almurisi
- Department of Pharmaceutical Technology, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Malaysia
| | - Khater Al-Japairai
- Department of Pharmaceutical Engineering, Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang, Gambang 26300, Malaysia
| | - Latifah Munirah Bakar
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM) Selangor, Shah Alam 40450, Malaysia
| | - Abd Almonem Doolaanea
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University College MAIWP International (UCMI), Kuala Lumpur 68100, Malaysia
| |
Collapse
|
14
|
Ojeda-Hernández DD, Hernández-Sapiéns MA, Reza-Zaldívar EE, Canales-Aguirre A, Matías-Guiu JA, Matías-Guiu J, Mateos-Díaz JC, Gómez-Pinedo U, Sancho-Bielsa F. Exosomes and Biomaterials: In Search of a New Therapeutic Strategy for Multiple Sclerosis. Life (Basel) 2022; 12:1417. [PMID: 36143453 PMCID: PMC9504193 DOI: 10.3390/life12091417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 02/07/2023] Open
Abstract
Current efforts to find novel treatments that counteract multiple sclerosis (MS) have pointed toward immunomodulation and remyelination. Currently, cell therapy has shown promising potential to achieve this purpose. However, disadvantages such as poor survival, differentiation, and integration into the target tissue have limited its application. A series of recent studies have focused on the cell secretome, showing it to provide the most benefits of cell therapy. Exosomes are a key component of the cell secretome, participating in the transfer of bioactive molecules. These nano-sized vesicles offer many therapeutical advantages, such as the capacity to cross the blood-brain barrier, an enrichable cargo, and a customizable membrane. Moreover, integrating of biomaterials into exosome therapy could lead to new tissue-specific therapeutic strategies. In this work, the use of exosomes and their integration with biomaterials is presented as a novel strategy in the treatment of MS.
Collapse
Affiliation(s)
- Doddy Denise Ojeda-Hernández
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC and Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Mercedes A. Hernández-Sapiéns
- Preclinical Evaluation Unit, Medical and Pharmaceutical Biotechnology Unit, CIATEJ-CONACyT, Guadalajara 44270, Mexico
| | - Edwin E. Reza-Zaldívar
- Tecnologico de Monterrey, The Institute for Obesity Research, Ave. General Ramón Corona 2514, Zapopan 45201, Mexico
| | - Alejandro Canales-Aguirre
- Preclinical Evaluation Unit, Medical and Pharmaceutical Biotechnology Unit, CIATEJ-CONACyT, Guadalajara 44270, Mexico
| | - Jordi A. Matías-Guiu
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC and Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Department of Neurology, Institute of Neurosciences, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | - Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Institute of Neurosciences, IdISSC and Hospital Clínico San Carlos, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Francisco Sancho-Bielsa
- Área de Fisiología, Departamento de Ciencias Médicas, Facultad de Medicina de Ciudad Real, UCLM, 13071 Ciudad Real, Spain
| |
Collapse
|
15
|
Ran N, Li W, Zhang R, Lin C, Zhang J, Wei Z, Li Z, Yuan Z, Wang M, Fan B, Shen W, Li X, Zhou H, Yao X, Kong X, Feng S. Autologous exosome facilitates load and target delivery of bioactive peptides to repair spinal cord injury. Bioact Mater 2022; 25:766-782. [PMID: 37056263 PMCID: PMC10086682 DOI: 10.1016/j.bioactmat.2022.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/30/2022] Open
Abstract
Spinal cord injury (SCI) causes motor, sensory and automatic impairment due to rarely axon regeneration. Developing effective treatment for SCI in the clinic is extremely challenging because of the restrictive axonal regenerative ability and disconnection of neural elements after injury, as well as the limited systemic drug delivery efficiency caused by blood spinal cord barrier. To develop an effective non-invasive treatment strategy for SCI in clinic, we generated an autologous plasma exosome (AP-EXO) based biological scaffold where AP-EXO was loaded with neuron targeting peptide (RVG) and growth-facilitating peptides (ILP and ISP). This scaffold can be targeted delivered to neurons in the injured area and elicit robust axon regrowth across the lesion core to the levels over 30-fold greater than naïve treatment, thus reestablish the intraspinal circuits and promote motor functional recovery after spinal cord injury in mice. More importantly, in ex vivo, human plasma exosomes (HP-EXO) loaded with combinatory peptides of RVG, ILP and ISP showed safety and no liver and kidney toxicity in the application to nude SCI mice. Combining the efficacy and safety, the AP-EXO-based personalized treatment confers functional recovery after SCI and showed immense promising in biomedical applications in treating SCI. It is helpful to expand the application of combinatory peptides and human plasma derived autologous exosomes in promoting regeneration and recovery upon SCI treatment.
Collapse
Affiliation(s)
- Ning Ran
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Wenxiang Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Renjie Zhang
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Caorui Lin
- Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Jianping Zhang
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhijian Wei
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghao Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Zhongze Yuan
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Min Wang
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Baoyou Fan
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenyuan Shen
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueying Li
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hengxing Zhou
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Yao
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaohong Kong
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Laboratory of Medical Molecular Virology, School of Medicine, Nankai University, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Tianjin Key Laboratory of Spine and Spinal Cord, National Spinal Cord Injury International Cooperation Base, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
- Corresponding author. Orthopedic Research Center of Shandong University &Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
16
|
Figueira RL, Antounians L, Zani-Ruttenstock E, Khalaj K, Zani A. Fetal lung regeneration using stem cell-derived extracellular vesicles: A new frontier for pulmonary hypoplasia secondary to congenital diaphragmatic hernia. Prenat Diagn 2022; 42:364-372. [PMID: 35191057 DOI: 10.1002/pd.6117] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 11/12/2022]
Abstract
The poor outcomes of babies with congenital diaphragmatic hernia (CDH) are directly related to pulmonary hypoplasia, a cosndition characterized by impaired lung development. Although the pathogenesis of pulmonary hypoplasia is not fully elucidated, there is now evidence that CDH patients have missing or dysregulated microRNAs (miRNAs) that regulate lung development. A prenatal therapy that supplements these missing/dysregulated miRNAs could be a strategy to rescue normal lung development. Extracellular vesicles (EVs), also known as exosomes when of small dimensions, are lipid-bound nanoparticles that can transfer their heterogeneous cargo (proteins, lipids, small RNAs) to target cells to induce biological responses. Herein, we review all studies that show evidence for stem cell-derived EVs as a regenerative therapy to rescue normal development in CDH fetal lungs. Particularly, we report studies showing that administration of EVs derived from amniotic fluid stem cells (AFSC-EVs) to models of pulmonary hypoplasia promotes fetal lung growth and maturation via transfer of miRNAs that are known to regulate lung developmental processes. We also describe that stem cell-derived EVs exert effects on vascular remodeling, thus possibly preventing postnatal pulmonary hypertension. Finally, we discuss future perspectives and challenges to translate this promising stem cell EV-based therapy to clinical practice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Rebeca Lopes Figueira
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Lina Antounians
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Elke Zani-Ruttenstock
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Kasra Khalaj
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Augusto Zani
- Developmental and Stem Cell Biology Program, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada.,Division of General and Thoracic Surgery, The Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada.,Department of Surgery, University of Toronto, Toronto, M5T 1P5, Canada
| |
Collapse
|
17
|
Li P, Lu X, Hu J, Dai M, Yan J, Tan H, Yu P, Chen X, Zhang C. Human amniotic fluid derived-exosomes alleviate hypoxic encephalopathy by enhancing angiogenesis in neonatal mice after hypoxia. Neurosci Lett 2022; 768:136361. [PMID: 34826550 DOI: 10.1016/j.neulet.2021.136361] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/11/2021] [Accepted: 11/19/2021] [Indexed: 12/17/2022]
Abstract
Neonatal hypoxic encephalopathy is a type of central nervous system dysfunction manifested by high mortality and morbidity. Exosomes play a crucial role in neuroprotection by enhancing angiogenesis. The objective of this study was to investigate the effect of human amniotic fluid-derived exosomes (hAFEXOs) on functional recovery in neonatal hypoxic encephalopathy. The transwell assay, scratch wound healing assay, and tube formation assay were used to evaluate the effect of hAFEXOs on the angiogenesis of human umbilical vein endothelial cells (HUVECs) after oxygen and glucose deprivation (OGD). The angiogenesis of microvascular endothelial cells (MECs) in the cortex was tested in neonatal mice treated with hAFEXOs or phosphate-buffered saline (PBS) after hypoxia. Expressions of hypoxia-inducible factor 1 α (HIF-1α) and vascular endothelial growth factor (VEGF) in the cerebral cortex were also tested by western blot. The Morris Water Maze Test (MWM) was carried out to detect the performance of spatial memory after processing with hAFEXOs or PBS. The results indicated that hAFEXOs favored tubing formation and migration of HUVECs after in vitro OGD. The hAFEXOs also favored the expression of CD31 in neonatal mice following hypoxia. The expressions of both HIF-1α and VEGF were significantly augmented in the cerebral cortex of neonatal mice which were treated with hAFEXOs. Moreover, the MWM test results showed that the performance of the spatial memory was better in the hAFEXO-treated group than in the PBS-treated group. Our study indicates that hAFEXOs alleviated hypoxic encephalopathy and enhanced angiogenesis in neonatal mice after hypoxia. In addition, hAFEXOs promoted migration and tube formation of HUVECs after OGD in vitro. These findings confirm that hAFEXOs show great potential for further studies aimed at developing therapeutic agents for hypoxic encephalopathy.
Collapse
Affiliation(s)
- Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha 410008, China; Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha 410008, China
| | - Xiaoxu Lu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiajia Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Minhui Dai
- Department of Clinical Dietitian, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jianqin Yan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Huiling Tan
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Peilin Yu
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xuliang Chen
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
18
|
Ohta M, Koshida S, Jimbo I, Oda M, Inoue R, Tsukahara T, Terahara M, Yanagi T, Nakahara S, Shibata M, Tsutsui H, Yoshida D, Furukawa O, Maruo Y. Chronological changes of serum exosome in preterm infants: A prospective study. Pediatr Int 2022; 64:e14933. [PMID: 34314566 DOI: 10.1111/ped.14933] [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: 04/07/2021] [Revised: 06/21/2021] [Accepted: 07/26/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Exosomes, which are observed in all human fluid, including serum, are nanosized extracellular vesicles with a mechanism of intercellular communication. Potential clinical applications of exosomes in neonatal diseases have recently been discussed. However, the characteristics of exosomes in serum during early infancy is unclear. METHODS In this prospective study, we evaluated the chronological changes in the concentration of serum-derived exosomes of 20 infants for 12 months after birth. RESULTS The average concentration of serum-derived exosomes was 4.6 × 1010 particles/mL at birth and increased significantly until the age of 48 weeks. There was a moderate correlation between the gestational age and the concentration of serum-derived exosomes both at birth (r = 0.54, P = 0.01) and during the 8 weeks after birth (r = 0.48, P < 0.001). A multivariable analysis showed that gestational age at birth was associated with the concentration of serum-derived exosomes at birth (partial regression coefficient, 0.86; 95% confidence interval, 0.37-1.37; P = 0.002). CONCLUSIONS The concentration of serum-derived exosomes in preterm infants increased both chronologically and by gestational age after birth. These basic data may help to further understand physiology of exosomes in preterm infants.
Collapse
Affiliation(s)
- Motoki Ohta
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shigeki Koshida
- Perinatal Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Itsuki Jimbo
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Japan
| | - Machi Oda
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Japan.,Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University - Hirakata Campus, Hirakata, Osaka, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Sakyo-ku, Kyoto, Japan.,Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University - Hirakata Campus, Hirakata, Osaka, Japan
| | | | - Masaki Terahara
- R&D Management Department, Meiji Co., Ltd, Hachiouji, Tokyo, Japan
| | - Takahide Yanagi
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Sayuri Nakahara
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masami Shibata
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Hidemi Tsutsui
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Daisuke Yoshida
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Ouki Furukawa
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
19
|
Ohta M, Koshida S, Jimbo I, Oda M, Inoue R, Tsukahara T, Terahara M, Nakamura Y, Maruo Y. Highest concentration of breast-milk-derived exosomes in colostrum. Pediatr Int 2022; 64:e15346. [PMID: 36370374 DOI: 10.1111/ped.15346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/08/2022] [Accepted: 08/31/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Exosomes are nanosized extracellular vesicles, that play important roles in intercellular immune regulation. They have potential therapeutic utility for neonatal diseases including necrotizing enterocolitis. Breast-milk-derived exosomes have recently shown beneficial effects on intestinal damage in vitro and in vivo. However, the chronological change in breast-milk-derived exosome concentrations after delivery are unclear. METHODS In this prospective study, we enrolled 17 mothers who delivered premature infants admitted to a neonatal intensive care unit in Japan. We measured the consecutive concentrations of breast-milk-derived exosomes in the mothers for 48 weeks after delivery. RESULTS The median concentration of breast-milk-derived exosomes was 1.62 × 108 particles/ml in colostrum, showing a significant decrease after 2 weeks (P < 0.01). There was no association between the exosome concentration in colostrum and maternal perinatal factors including parity, mode of delivery, maternal age, and gestational age at delivery. CONCLUSIONS We concluded that breast-milk-derived exosomes were the richest in colostrum. Our basic data regarding breast-milk-derived exosomes are expected to aid in the clinical application of exosomes for treating neonatal diseases.
Collapse
Affiliation(s)
- Motoki Ohta
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shigeki Koshida
- Perinatal Center, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Itsuki Jimbo
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan
| | - Machi Oda
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan.,Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University - Hirakata Campus, Hirakata, Osaka, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Agricultural and Life Sciences, Kyoto Prefectural University, Kyoto, Japan.,Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University - Hirakata Campus, Hirakata, Osaka, Japan
| | | | - Masaki Terahara
- R&D Management Department, Meiji Co., Ltd., Hachiouji, Tokyo, Japan
| | - Yoshitaka Nakamura
- Food Microbiology and Function Research Laboratories, Meiji Co., Ltd., Hachiouji, Tokyo, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
20
|
Balsamo F, Tian Y, Pierro A, Li B. Amniotic fluid stem cells: A novel treatment for necrotizing enterocolitis. Front Pediatr 2022; 10:1020986. [PMID: 36533245 PMCID: PMC9751649 DOI: 10.3389/fped.2022.1020986] [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: 08/16/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a gastrointestinal disease frequently prevalent in premature neonates. Despite advances in research, there is a lack of accurate, early diagnoses of NEC and the current therapeutic approaches remain exhausted and disappointing. In this review, we have taken a close look at the regenerative medical literature available in the context of NEC treatment. Stem cells from amniotic fluid (AFSC) administration may have the greatest protective and restorative effects on NEC. This review summarizes the potential protection and restoration AFSCs have on NEC-induced intestinal injury while comparing various components within AFSCs like conditioned medium (CM) and extracellular vesicles (EVs). In addition to therapeutic interventions that focus on targeting intestinal epithelial damage and regeneration, a novel discovery that AFSCs act in a Wnt-dependent manner provides insight into this mechanism of protection. Finally, we have highlighted the most important aspects that remain unknown that should be considered to guide future research on the translational application of AFSC-based therapy. We hope that this will be a beneficial frame of reference for the guidance of future studies and towards the clinical application of AFSC and/or its derivatives as a treatment against NEC.
Collapse
Affiliation(s)
- Felicia Balsamo
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yina Tian
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bo Li
- Division of General and Thoracic Surgery, Translational Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
21
|
Chand K, Nano R, Wixey J, Patel J. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:372-382. [PMID: 35485440 PMCID: PMC9052430 DOI: 10.1093/stcltm/szac005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/12/2021] [Indexed: 11/25/2022] Open
Abstract
Fetal growth restriction (FGR) occurs when a fetus is unable to grow normally due to inadequate nutrient and oxygen supply from the placenta. Children born with FGR are at high risk of lifelong adverse neurodevelopmental outcomes, such as cerebral palsy, behavioral issues, and learning and attention difficulties. Unfortunately, there is no treatment to protect the FGR newborn from these adverse neurological outcomes. Chronic inflammation and vascular disruption are prevalent in the brains of FGR neonates and therefore targeted treatments may be key to neuroprotection. Tissue repair and regeneration via stem cell therapies have emerged as a potential clinical intervention for FGR babies at risk for neurological impairment and long-term disability. This review discusses the advancement of research into stem cell therapy for treating neurological diseases and how this may be extended for use in the FGR newborn. Leading preclinical studies using stem cell therapies in FGR animal models will be highlighted and the near-term steps that need to be taken for the development of future clinical trials.
Collapse
Affiliation(s)
- Kirat Chand
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Rachel Nano
- Cancer and Ageing Research Program, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Julie Wixey
- Julie Wixey, Faculty of Medicine, Royal Brisbane and Women’s Hospital, The University of Queensland Centre for Clinical Research, Herston 4029 QLD, Australia.
| | - Jatin Patel
- Corresponding authors: Jatin Patel, Translational Research Institute, Queensland University of Technology, 37 Kent Street, Woolloongabba 4102 QLD, Australia.
| |
Collapse
|
22
|
Filip R. An Update on the Role of Extracellular Vesicles in the Pathogenesis of Necrotizing Enterocolitis and Inflammatory Bowel Diseases. Cells 2021; 10:cells10113202. [PMID: 34831425 PMCID: PMC8622309 DOI: 10.3390/cells10113202] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022] Open
Abstract
Some of the most fundamental influences of microorganisms inhabiting the human intestinal tract are exerted during infant development and impact the maturation of intestinal mucosa and gut immune system. The impact of bacteria on the host gut immune system is partially mediated via released extracellular vesicles (EVs). The heterogeneity in EV content, size, and bacterial species origin can have an impact on intestinal cells, resulting in inflammation and an immune response, or facilitate pathogen entry into the gut wall. In mammals, maintaining the integrity of the gut barrier might also be an evolutionary function of maternal milk EVs. Recently, the usage of EVs has been explored as a novel therapeutic approach in several pathological conditions, including necrotizing enterocolitis (NEC) and inflammatory bowel disease (IBD). In this review, we attempt to summarize the current knowledge of EV biology, followed by a discussion of the role that EVs play in gut maturation and the pathogenesis of NEC and IBD.
Collapse
Affiliation(s)
- Rafał Filip
- Department of Gastroenterology with IBD, Unit of Clinical Hospital 2 in Rzeszow, Lwowska 60, 35-310 Rzeszow, Poland;
- Faculty of Medicine, University of Rzeszow, Aleja Majora Wacława Kopisto 2a, 35-210 Rzeszow, Poland
| |
Collapse
|
23
|
Human Umbilical Cord Mesenchymal Stem Cell-Derived Exosomes Attenuate Oxygen-Glucose Deprivation/Reperfusion-Induced Microglial Pyroptosis by Promoting FOXO3a-Dependent Mitophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6219715. [PMID: 34765084 PMCID: PMC8577931 DOI: 10.1155/2021/6219715] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 12/17/2022]
Abstract
Background Mesenchymal stem cell-derived exosomes (MSC-exos) have been recognized as a promising therapeutic strategy for neonatal hypoxic-ischemic brain damage (HIBD). Recently, microglial pyroptosis was shown to play a vital role in the progression of neonatal HIBD. However, whether MSC-exos improve HIBD by regulating microglial pyroptosis remains unknown. Methods Exosomes were isolated from human umbilical cord mesenchymal stem cells (huMSCs) and identified by transmission electron microscopy (TEM), western blot, and nanoparticle tracking analysis (NTA). BV-2 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce microglial ischemia/reperfusion (I/R) in vitro. CCK-8, ELISA, western blot, and Hoechst 33342/PI double staining were performed to detect the pyroptosis of BV-2 cells. Conditioned medium (CM) from BV-2 cells exposed to different treatments was used to investigate its effect on neuronal injury. Moreover, 3-methyladenine (3-MA) and mitochondrial division inhibitor-1 (mdi-1) were used to verify the involvement of mitophagy in the protection of MSC-exos against OGD/R-induced pyroptosis in BV-2 cells. Finally, FOXO3a siRNA was used to investigate the involvement of FOXO3a in MSC-exo-induced mitophagy and pyroptosis inhibition. Results Exosomes from huMSCs were successfully extracted. In OGD/R-exposed BV-2 cells, MSC-exos increased cell viability and decreased the expression of NLRP3, cleaved caspase-1, and GSDMD-N as well as the release of IL-1β and IL-18. Compared with CM from OGD/R-exposed BV-2 cells treated with PBS, CM from OGD/R-exposed BV-2 cells treated with MSC-exos significantly increased the viability of SH-SY5Y cells and decreased LDH release. MSC-exos also increased the expression of TOM20 and COX IV in OGD/R-exposed BV-2 cells. Additionally, 3-MA and mdi-1 attenuated the inhibition of pyroptosis with MSC-exo treatment. Furthermore, FOXO3a siRNA partially abolished the neuroprotective effect of MSC-exos and attenuated mitophagy and pyroptosis inhibition induced by MSC-exo treatment. Conclusions Our findings demonstrated that MSC-exos increased FOXO3a expression to enhance mitophagy, therefore protecting microglia from I/R-induced pyroptosis and alleviating subsequent neuronal injury.
Collapse
|
24
|
Matheakakis A, Batsali A, Papadaki HA, Pontikoglou CG. Therapeutic Implications of Mesenchymal Stromal Cells and Their Extracellular Vesicles in Autoimmune Diseases: From Biology to Clinical Applications. Int J Mol Sci 2021; 22:10132. [PMID: 34576296 PMCID: PMC8468750 DOI: 10.3390/ijms221810132] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are perivascular multipotent stem cells originally identified in the bone marrow (BM) stroma and subsequently in virtually all vascularized tissues. Because of their ability to differentiate into various mesodermal lineages, their trophic properties, homing capacity, and immunomodulatory functions, MSCs have emerged as attractive candidates in tissue repair and treatment of autoimmune disorders. Accumulating evidence suggests that the beneficial effects of MSCs may be primarily mediated via a number of paracrine-acting soluble factors and extracellular vesicles (EVs). EVs are membrane-coated vesicles that are increasingly being acknowledged as playing a key role in intercellular communication via their capacity to carry and deliver their cargo, consisting of proteins, nucleic acids, and lipids to recipient cells. MSC-EVs recapitulate the functions of the cells they originate, including immunoregulatory effects but do not seem to be associated with the limitations and concerns of cell-based therapies, thereby emerging as an appealing alternative therapeutic option in immune-mediated disorders. In the present review, the biology of MSCs will be outlined and an overview of their immunomodulatory functions will be provided. In addition, current knowledge on the features of MSC-EVs and their immunoregulatory potential will be summarized. Finally, therapeutic applications of MSCs and MSC-EVs in autoimmune disorders will be discussed.
Collapse
Affiliation(s)
- Angelos Matheakakis
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Aristea Batsali
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Helen A. Papadaki
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| | - Charalampos G. Pontikoglou
- Department of Hematology, School of Medicine, University of Crete, 71500 Heraklion, Greece; (A.M.); (H.A.P.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71500 Heraklion, Greece;
| |
Collapse
|
25
|
Mizenko RR, Brostoff T, Rojalin T, Koster HJ, Swindell HS, Leiserowitz GS, Wang A, Carney RP. Tetraspanins are unevenly distributed across single extracellular vesicles and bias sensitivity to multiplexed cancer biomarkers. J Nanobiotechnology 2021; 19:250. [PMID: 34419056 PMCID: PMC8379740 DOI: 10.1186/s12951-021-00987-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023] Open
Abstract
Background Tetraspanin expression of extracellular vesicles (EVs) is often used as a surrogate for their detection and classification, a practice that typically assumes their consistent expression across EV sources. Results Here we demonstrate that there are distinct patterns in colocalization of tetraspanin expression of EVs enriched from a variety of in vitro and in vivo sources. We report an optimized method for the use of single particle antibody-capture and fluorescence detection to identify subpopulations according to tetraspanin expression and compare our findings with nanoscale flow cytometry. We found that tetraspanin profile is consistent from a given EV source regardless of isolation method, but that tetraspanin profiles are distinct across various sources. Tetraspanin profiles measured by flow cytometry do not totally agree, suggesting that limitations in subpopulation detection significantly impact apparent protein expression. We further analyzed tetraspanin expression of single EVs captured non-specifically, revealing that tetraspanin capture can bias the apparent multiplexed tetraspanin profile. Finally, we demonstrate that this bias can have significant impact on diagnostic sensitivity for tumor-associated EV surface markers. Conclusion Our findings may reveal key insights into protein expression heterogeneity of EVs that better inform EV capture and detection platforms for diagnostic or other downstream use. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00987-1.
Collapse
Affiliation(s)
- Rachel R Mizenko
- Department of Biomedical Engineering, University of California, Davis, USA
| | - Terza Brostoff
- Department of Pathology, University of California, San Diego, USA
| | - Tatu Rojalin
- Department of Biomedical Engineering, University of California, Davis, USA
| | - Hanna J Koster
- Department of Biomedical Engineering, University of California, Davis, USA
| | | | - Gary S Leiserowitz
- Division of Gynecologic Oncology, University of California Davis Medical Center, Sacramento, CA, USA
| | - Aijun Wang
- Department of Biomedical Engineering, University of California, Davis, USA.,Department of Surgery, University of California, Davis, USA
| | - Randy P Carney
- Department of Biomedical Engineering, University of California, Davis, USA.
| |
Collapse
|
26
|
Gupta A, Maffulli N, Rodriguez HC, Mistovich RJ, Delfino K, Cady C, Fauser AM, Cundiff ED, Martinez MA, Potty AG. Cell-free stem cell-derived extract formulation for treatment of knee osteoarthritis: study protocol for a preliminary non-randomized, open-label, multi-center feasibility and safety study. J Orthop Surg Res 2021; 16:514. [PMID: 34416898 PMCID: PMC8377854 DOI: 10.1186/s13018-021-02672-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Musculoskeletal conditions are highly prevalent, and knee OA is most common. Current treatment modalities have limitations and either fail to solve the underlying pathophysiology or are highly invasive. To address these limitations, attention has focused on the use of biologics. The efficacy of these devices is attributed to presence of growth factors (GFs), cytokines (CKs), and extracellular vesicles (EVs). With this in mind, we formulated a novel cell-free stem cell-derived extract (CCM) from human progenitor endothelial stem cells (hPESCs). A preliminary study demonstrated the presence of essential components of regenerative medicine, namely GFs, CKs, and EVs, including exosomes, in CCM. The proposed study aims to evaluate the safety and efficacy of intraarticular injection of the novel cell-free stem cell-derived extract (CCM) for the treatment of knee OA. METHODS AND ANALYSIS This is a non-randomized, open-label, multi-center, prospective study in which the safety and efficacy of intraarticular CCM in patients suffering from grade II/III knee OA will be evaluated. Up to 20 patients with grade II/III OA who meet the inclusion and exclusion criteria will be consented and screened to recruit 12 patients to receive treatment. The study will be conducted at up to 2 sites within the USA, and the 12 participants will be followed for 24 months. The study participants will be monitored for adverse reactions and assessed using Numeric Pain Rating Scale (NPRS), Patient-Reported Outcomes Measurement Information System (PROMIS) Score, Knee Injury and Osteoarthritis Outcome Score Jr. (KOOS Jr.), 36-ietm short form survey (SF-36), Single Assessment Numeric Evaluation (SANE), physical exams, plain radiography, and magnetic resonance imaging (MRI) with Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score for improvements in pain, function, satisfaction, and cartilage regeneration. DISCUSSION This prospective study will provide valuable information into the safety and efficacy of intraarticular administration of cell-free stem cell-derived extract (CCM) in patients suffering with grade II/III knee OA. The outcomes from this initial study of novel CCM will lay the foundation for a larger randomized, placebo-controlled, multi-center clinical trial of intraarticular CCM for symptomatic knee OA. TRIAL REGISTRATION Registered on July 21, 2021. ClinicalTrials.gov NCT04971798.
Collapse
Affiliation(s)
- Ashim Gupta
- General Therapeutics, 2956 Washington Blvd, Cleveland Heights, OH 44118 USA
- Future Biologics, Lawrenceville, GA USA
- South Texas Orthopedic Research Institute (STORI Inc.), Laredo, TX USA
- Veterans in Pain (V.I.P.), Los Angeles, CA USA
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Orthopedica” Department, Hospital of Salerno, Salerno, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent, UK
| | - Hugo C. Rodriguez
- Future Biologics, Lawrenceville, GA USA
- South Texas Orthopedic Research Institute (STORI Inc.), Laredo, TX USA
- School of Osteopathic Medicine, University of The Incarnate Word, San Antonio, TX USA
- Future Physicians of South Texas, San Antonio, TX USA
| | - R. Justin Mistovich
- General Therapeutics, 2956 Washington Blvd, Cleveland Heights, OH 44118 USA
- Department of Orthopaedics, School of Medicine, Case Western Reserve University, Cleveland, OH USA
| | - Kristin Delfino
- Southern Illinois University, School of Medicine, Springfield, IL USA
| | - Craig Cady
- General Therapeutics, 2956 Washington Blvd, Cleveland Heights, OH 44118 USA
- Bohlander Stem Cell Research Laboratory, Department of Biology, Bradley University, Peoria, IL USA
| | - Anne-Marie Fauser
- Bohlander Stem Cell Research Laboratory, Department of Biology, Bradley University, Peoria, IL USA
| | - Echo D. Cundiff
- General Therapeutics, 2956 Washington Blvd, Cleveland Heights, OH 44118 USA
| | | | - Anish G. Potty
- General Therapeutics, 2956 Washington Blvd, Cleveland Heights, OH 44118 USA
- South Texas Orthopedic Research Institute (STORI Inc.), Laredo, TX USA
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London, UK
- Laredo Sports Medicine Clinic, Laredo, TX USA
| |
Collapse
|
27
|
Zani-Ruttenstock E, Antounians L, Khalaj K, Figueira RL, Zani A. The Role of Exosomes in the Treatment, Prevention, Diagnosis, and Pathogenesis of COVID-19. Eur J Pediatr Surg 2021; 31:326-334. [PMID: 34161984 DOI: 10.1055/s-0041-1731294] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), continues to be a major health concern. In search for novel treatment strategies against COVID-19, exosomes have attracted the attention of scientists and pharmaceutical companies worldwide. Exosomes are small extracellular vesicles, secreted by all types of cells, and considered as key mediators of intercellular communication and stem-cell paracrine signaling. Herein, we reviewed the most recent literature about the role of exosomes as potential agents for treatment, prevention, diagnosis, and pathogenesis of COVID-19. Several studies and ongoing clinical trials have been investigating the anti-inflammatory, immunomodulatory, and reparative effects of exosomes derived from mesenchymal stem/stromal cells for COVID-19-related acute lung injury. Other studies reported that exosomes play a key role in convalescent plasma therapy for COVID-19, and that they could be of use for the treatment of COVID-19 Kawasaki's-like multisystem inflammatory syndrome and as drug delivery nanocarriers for antiviral therapy. Harnessing some advantageous aspects of exosome biology, such as their endogenous origin, capability of crossing biological barriers, high stability in circulation, and low toxicity and immunogenicity, several companies have been testing exosome-based vaccines against SARS-CoV-2. As they carry cargos that mimic the status of parent cells, exosomes can be isolated from a variety of sources, including plasma, and employed as biomarkers of COVID-19. Lastly, there is growing evidence supporting the role of exosomes in COVID-19 infection, spread, reactivation, and reinfection. The lessons learned using exosomes for COVID-19 will help determine their efficacy and applicability in other clinical conditions.
Collapse
Affiliation(s)
- Elke Zani-Ruttenstock
- Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lina Antounians
- Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kasra Khalaj
- Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rebeca L Figueira
- Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Augusto Zani
- Division of General and Thoracic Surgery, Hospital for Sick Children, Toronto, Ontario, Canada.,Developmental and Stem Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
28
|
A review of the role of extracellular vesicles in neonatal physiology and pathology. Pediatr Res 2021; 90:289-299. [PMID: 33184501 DOI: 10.1038/s41390-020-01240-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 12/23/2022]
Abstract
Extracellular vesicles (EVs) are cell-derived membrane-bound particles, extensively investigated across many fields to improve the understanding of pathophysiological processes, as biomarkers of disease and as therapeutic targets for pharmacological intervention. We aim to describe the current knowledge of EVs detected in the body fluids of human neonates, both term and preterm, from birth to 4 weeks of age. To date, EVs have been described in several neonatal body fluids, including cerebrospinal fluid, umbilical cord blood, neonatal blood, tracheal aspirates and urine. These studies demonstrate some important roles of EVs in the neonatal population, particularly in haemostasis. Moreover, some studies have demonstrated the pathophysiological mechanisms and the identification of potential biomarkers of neonatal disease. We must continue to build on this knowledge, evaluating the role of EVs in neonatal pathology, particularly in prematurity and during the perinatal adaption period. Future studies should use larger numbers, robust EV characterisation techniques and always correlate the findings to clinical outcomes. IMPACT: This article summarises the current knowledge of the effect of EVs in neonates. It describes the potential compensatory role of EVs in neonatal haemostasis. It also describes the role of EVs as mediators of pathology and as potential biomarkers of perinatal and neonatal disease.
Collapse
|
29
|
Chen Y, Yuan X, Li Y, Chen J, Wu S, Jiang A, Miao X, Shu Q. Circulating exosomal microRNA-18a-5p accentuates intestinal inflammation in Hirschsprung-associated enterocolitis by targeting RORA. Am J Transl Res 2021; 13:4182-4196. [PMID: 34150007 PMCID: PMC8205693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/15/2021] [Indexed: 06/12/2023]
Abstract
The relevance of stem cell-derived exosomes has been implicated in necrotizing enterocolitis, while the involvement of serum-derived exosomes from children with Hirschsprung-associated enterocolitis (HAEC) in pathogenesis of HAEC remains unclear. This study set to identify the roles of exosomal microRNA (miR)-18a-5p from sera of HAEC patients in human-derived colonic epithelial NCM460 cells and in mice with HAEC. Exosomes were isolated from the sera of healthy children (Healthy-exo), patients with Hirschsprung's disease (HSCR) (HSCR-exo) or HAEC (HAEC-exo). A microarray analysis of miRNAs was implemented to assess the enrichment of miRNAs in these exosomes. HAEC-exo was significantly enriched in miR-18a-5p. HAEC-exo led to the generation of a pro-inflammatory microenvironment, inhibition of cellular DNA synthesis, and promotion of apoptosis in NCM460 cells. Mechanistically, miR-18a-5p targeted and repressed retinoid-related orphan receptor α (RORA) expression, thereby regulating the Sirtuin 1 (SIRT1)/nuclear factor-kappa B (NFκB) pathway. Overexpression of RORA ameliorated inflammatory damage in NCM460 cells caused by exosomal miR-18a-5p. HAEC-exo exacerbated inflammatory damage in HAEC mice, and this facilitation was reversed after RORA overexpression. Collectively, exosomal miR-18a-5p was a promoter of HAEC, which induces the intestine cell apoptosis and inflammatory responses through the inhibition of SIRT1/NFκB pathway by targeting RORA.
Collapse
Affiliation(s)
- Yi Chen
- Department of Nosocomial Infection, Yiwu Maternity and Children HospitalJinhua 322000, Zhejiang, P. R. China
| | - Xiaojian Yuan
- Department of Pediatrics, Zhejiang University School of MedicineHangzhou 310011, Zhejiang, P. R. China
| | - Yonglin Li
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Jie Chen
- Department of Pediatric Surgery, Jiaxing Maternal and Child Health HospitalJiaxing 314051, Zhejiang, P. R. China
| | - Shannan Wu
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Amin Jiang
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Xuefeng Miao
- Department of Pediatric Surgery, Yiwu Maternity and Child Health HospitalJinhua 322000, Zhejiang, P. R. China
| | - Qiang Shu
- Department of Cardio-Thoracic Surgery, Children’s Hospital Affiliated to Zhejiang UniversityHangzhou 310000, Zhejiang, P. R. China
| |
Collapse
|
30
|
Administration of extracellular vesicles derived from human amniotic fluid stem cells: a new treatment for necrotizing enterocolitis. Pediatr Surg Int 2021; 37:301-309. [PMID: 33566163 DOI: 10.1007/s00383-020-04826-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/30/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease. Amniotic fluid stem cells (AFSC) improve NEC injury but human translation remains difficult. We aimed to evaluate the use of extracellular vesicles (EV) derived from human AFSC. METHODS Human AFSC (hAFSC) were cultured according to the protocol (Celprogen Inc., California, U.S.A.). Conditioned medium was obtained, ultra-centrifuged, and EV were suspended in phosphate-buffered saline (PBS). C57BL/6 pups were grouped into: (1) breast-fed (Control, n = 11); (2) NEC + placebo (NEC + PBS; n = 10); and (3) NEC + treatment (NEC + EV; n = 11). NEC was induced post-natal days P5-9 by (A) gavage feeding hyperosmolar formula; (B) hypoxia for 10 min; and (C) lipopolysaccharide. Intra-peritoneal injections of PBS or hAFSC-EV were given on P6-7. All animals were sacrificed on P9 and terminal ileum harvested. RESULTS hAFSC-EV administration reduced intestinal injury (p = 0.0048), NEC incidence (score ≥ 2), and intestinal inflammation (IL-6 p < 0.0001; TNF-α p < 0.0001). Intestinal stem cell expression (Lgr5 +) and cellular proliferation (Ki67) were enhanced above control levels following hAFSC-EV administration (Lgr5 p = 0.0003; Ki67 p < 0.0001). CONCLUSION hAFSC-EV administration reduced intestinal NEC injury and inflammation while increasing stem cell expression and cellular proliferation. hAFSC-EV administration may induce similar beneficial effects to exogenous stem cells.
Collapse
|
31
|
Gupta A, Cady C, Fauser AM, Rodriguez HC, Mistovich RJ, Potty AGR, Maffulli N. Cell-free Stem Cell-Derived Extract Formulation for Regenerative Medicine Applications. Int J Mol Sci 2020; 21:9364. [PMID: 33316880 PMCID: PMC7763336 DOI: 10.3390/ijms21249364] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 11/20/2020] [Accepted: 12/05/2020] [Indexed: 12/17/2022] Open
Abstract
Stem cells for regenerative medicine purposes offer therapeutic benefits, but disadvantages are still ill defined. The benefit of stem cells may be attributed to their secretion of growth factors (GFs), cytokines (CKs), and extracellular vesicles (EVs), including exosomes. We present a novel cell-free stem cell-derived extract (CCM), formulated from human progenitor endothelial stem cells (hPESCs), characterized for biologically active factors using ELISA, nanoparticle tracking analysis and single particle interferometric reflectance imaging sensing. The effect on fibroblast proliferation and ability to induce stem cell migration was analyzed using Alamar Blue proliferation and Transwell migration assays, respectively. GFs including IGFBP 1, 2, 3, and 6, insulin, growth hormone, PDGF-AA, TGF-α, TGF-β1, VEGF, and the anti-inflammatory cytokine, IL-1RA were detected. Membrane enclosed particles within exosome size range and expressing exosome tetraspanins CD81 and CD9 were identified. CCM significantly increased cell proliferation and induced stem cell migration. Analysis of CCM revealed presence of GFs, CKs, and EVs, including exosomes. The presence of multiple factors including exosomes within one formulation, the ability to promote cell proliferation and induce stem cell migration may reduce inflammation and pain, and augment tissue repair.
Collapse
Affiliation(s)
- Ashim Gupta
- General Therapeutics, Cleveland Heights, OH 44118, USA; (A.G.); (C.C.); (R.J.M.); (A.G.R.P.)
- Future Biologics, Lawrenceville, GA 30043, USA;
- BioIntegrate, Lawrenceville, GA 30043, USA
- South Texas Orthopaedic Research Institute, Laredo, TX 78045, USA
- Veterans in Pain, Valencia, CA 91354, USA
| | - Craig Cady
- General Therapeutics, Cleveland Heights, OH 44118, USA; (A.G.); (C.C.); (R.J.M.); (A.G.R.P.)
- Bohlander Stem Cell Research Laboratory, Department of Biology, Bradley University, Peoria, IL 61625, USA;
| | - Anne-Marie Fauser
- Bohlander Stem Cell Research Laboratory, Department of Biology, Bradley University, Peoria, IL 61625, USA;
| | - Hugo C. Rodriguez
- Future Biologics, Lawrenceville, GA 30043, USA;
- South Texas Orthopaedic Research Institute, Laredo, TX 78045, USA
- School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
- Future Physicians of South Texas, San Antonio, TX 78235, USA
| | - R. Justin Mistovich
- General Therapeutics, Cleveland Heights, OH 44118, USA; (A.G.); (C.C.); (R.J.M.); (A.G.R.P.)
- Department of Orthopaedics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Anish G. R. Potty
- General Therapeutics, Cleveland Heights, OH 44118, USA; (A.G.); (C.C.); (R.J.M.); (A.G.R.P.)
- South Texas Orthopaedic Research Institute, Laredo, TX 78045, USA
- School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
- Laredo Sports Medicine Clinic, Laredo, TX 78041, USA
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, 84084 Fisciano, Italy
- San Giovanni di Dio e Ruggi D’Aragona Hospital “Clinica Orthopedica” Department, Hospital of Salerno, 84124 Salerno, Italy
- Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, Queen Mary University of London, London E1 4DG, UK
- School of Pharmacy and Bioengineering, Keele University School of Medicine, Stoke on Trent ST5 5BG, UK
| |
Collapse
|
32
|
O'Reilly D, Dorodnykh D, Avdeenko NV, Nekliudov NA, Garssen J, Elolimy AA, Petrou L, Simpson MR, Yeruva L, Munblit D. Perspective: The Role of Human Breast-Milk Extracellular Vesicles in Child Health and Disease. Adv Nutr 2020; 12:59-70. [PMID: 32838428 PMCID: PMC7849950 DOI: 10.1093/advances/nmaa094] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/06/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
Human breast milk (HM) contains multiple bioactive substances determining its impact on children's health. Extracellular vesicles (EVs) are a heterogeneous group of secreted nanoparticles that are present in HM and may be partially responsible for its beneficial effects. The precise roles and content of EVs in HM remain largely unknown. To examine this, we performed a short narrative review on the literature focusing on HM EVs to contextualize the available data, followed by a scoping review of MEDLINE and Embase databases. We identified 424 nonduplicate citations with 19 original studies included. In this perspective, we summarize the evidence around HM EVs, highlight some theoretical considerations based on existing evidence, and provide an overview of some challenges associated with the complexity and heterogeneity of EV research. We consider how the existing data from HM studies conform to the minimal information for studies of EVs (MISEV) guidelines. Across the studies a variety of research methods were utilized involving both bench-based and translational methods, and a range of different EV contents were examined including RNA, proteins, and glycopeptides. We observed a variety of health outcomes in these studies, including allergy and atopy, necrotizing enterocolitis, and HIV. While some promising results have been demonstrated, the heterogeneity in outcomes of interest, methodological limitations, and relatively small number of studies in the field make comparison between studies or further translational work problematic. To date, no studies have examined normative values of HM EVs in a large, diverse population or with respect to potentially important influencing factors such as timing (hind- vs. foremilk), stage (colostrum vs. mature milk), and infant age (preterm vs. term), which makes extrapolation from bench or "basic" research impossible. Future research should focus on addressing the current inadequacies in the literature and utilize MISEV guidelines to inform study design.
Collapse
Affiliation(s)
| | - Denis Dorodnykh
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nina V Avdeenko
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Nikita A Nekliudov
- Department of Pediatrics and Pediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ahmed A Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Loukia Petrou
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway,Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
| | - Laxmi Yeruva
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA,Arkansas Children's Nutrition Center, Little Rock, AR, USA,Arkansas Children's Research Institute, Little Rock, AR, USA
| | | |
Collapse
|
33
|
Ramiro-Cortijo D, Singh P, Liu Y, Medina-Morales E, Yakah W, Freedman SD, Martin CR. Breast Milk Lipids and Fatty Acids in Regulating Neonatal Intestinal Development and Protecting against Intestinal Injury. Nutrients 2020; 12:E534. [PMID: 32092925 PMCID: PMC7071444 DOI: 10.3390/nu12020534] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/14/2020] [Accepted: 02/16/2020] [Indexed: 12/13/2022] Open
Abstract
Human breast milk is the optimal source of nutrition for infant growth and development. Breast milk fats and their downstream derivatives of fatty acids and fatty acid-derived terminal mediators not only provide an energy source but also are important regulators of development, immune function, and metabolism. The composition of the lipids and fatty acids determines the nutritional and physicochemical properties of human milk fat. Essential fatty acids, including long-chain polyunsaturated fatty acids (LCPUFAs) and specialized pro-resolving mediators, are critical for growth, organogenesis, and regulation of inflammation. Combined data including in vitro, in vivo, and human cohort studies support the beneficial effects of human breast milk in intestinal development and in reducing the risk of intestinal injury. Human milk has been shown to reduce the occurrence of necrotizing enterocolitis (NEC), a common gastrointestinal disease in preterm infants. Preterm infants fed human breast milk are less likely to develop NEC compared to preterm infants receiving infant formula. Intestinal development and its physiological functions are highly adaptive to changes in nutritional status influencing the susceptibility towards intestinal injury in response to pathological challenges. In this review, we focus on lipids and fatty acids present in breast milk and their impact on neonatal gut development and the risk of disease.
Collapse
Affiliation(s)
- David Ramiro-Cortijo
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Yan Liu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - Esli Medina-Morales
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
| | - William Yakah
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
| | - Steven D. Freedman
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; (D.R.-C.); (P.S.); (Y.L.); (E.M.-M.); (S.D.F.)
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Camilia R. Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA;
- Division of Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
34
|
Gupta A, El-Amin SF, Levy HJ, Sze-Tu R, Ibim SE, Maffulli N. Umbilical cord-derived Wharton's jelly for regenerative medicine applications. J Orthop Surg Res 2020; 15:49. [PMID: 32054483 PMCID: PMC7017504 DOI: 10.1186/s13018-020-1553-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/09/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The last decade has seen an explosion in the interest in using biologics for regenerative medicine applications, including umbilical cord-derived Wharton's Jelly. There is insufficient literature assessing the amount of growth factors, cytokines, hyaluronic acid, and extracellular vesicles including exosomes in these products. The present study reports the development of a novel Wharton's jelly formulation and evaluates the presence of growth factors, cytokines, hyaluronic acid, and extracellular vesicles including exosomes. METHODS Human umbilical cords were obtained from consenting caesarian section donors. The Wharton's jelly was then isolated from the procured umbilical cord and formulated into an injectable form. Randomly selected samples from different batches were analyzed for sterility testing and to quantify the presence of growth factors, cytokines, hyaluronic acid, and extracellular vesicles. RESULTS All samples passed the sterility test. Growth factors including IGFBP 1, 2, 3, 4, and 6, TGF-α, and PDGF-AA were detected. Several immunomodulatory cytokines, such as RANTES, IL-6R, and IL-16, were also detected. Pro-inflammatory cytokines MCSFR, MIP-1a; anti-inflammatory cytokines TNF-RI, TNF-RII, and IL-1RA; and homeostatic cytokines TIMP-1 and TIMP-2 were observed. Cytokines associated with wound healing, ICAM-1, G-CSF, GDF-15, and regenerative properties, GH, were also expressed. High concentrations of hyaluronic acid were observed. Particles in the extracellular vesicle size range were also detected and were enclosed by the membrane, indicative of true extracellular vesicles. CONCLUSION There are numerous growth factors, cytokines, hyaluronic acid, and extracellular vesicles present in the Wharton's jelly formulation analyzed. The amount of these factors in Wharton's jelly is higher compared with other biologics and may play a role in reducing inflammation and pain and augment healing of musculoskeletal injuries.
Collapse
Affiliation(s)
- Ashim Gupta
- BioIntegrate, New York, NY USA
- South Texas Orthopaedic Research Institute, Laredo, TX USA
- Department of Psychology, Illinois Wesleyan University, Bloomington, IL USA
- Future Biologics, Lawrenceville, GA USA
| | - Saadiq F. El-Amin
- BioIntegrate, New York, NY USA
- El-Amin Orthopaedic and Sports Medicine Institute, Duluth, GA USA
| | - Howard J. Levy
- BioIntegrate, New York, NY USA
- Department of Orthopaedic Surgery, Lenox Hill Hospital, Northwell Health, New York, NY USA
| | - Rebecca Sze-Tu
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | | | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Fisciano, Italy
- Queen Mary University of London Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, London, England
- Keele University Faculty of Medicine, School of Pharmacy and Bioengineering, Stoke on Trent, England
| |
Collapse
|
35
|
Liu M, Qiu Y, Xue Z, Wu R, Li J, Niu X, Yuan J, Wang Y, Wu Q. Small extracellular vesicles derived from embryonic stem cells restore ovarian function of premature ovarian failure through PI3K/AKT signaling pathway. Stem Cell Res Ther 2020; 11:3. [PMID: 31900201 PMCID: PMC6942273 DOI: 10.1186/s13287-019-1508-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/18/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Premature ovarian failure (POF) has a great impact on reproductive endocrine function in females, and it is an important cause of infertility. Previous studies have demonstrated that small extracellular vesicles (sEVs) derived from stem cells play an important role in tissue regeneration. This study aimed to investigate the therapeutic effect of sEVs derived from embryonic stem cells (ESCs-sEVs) on damaged ovaries and explore the underlying molecular mechanisms. METHODS Mice POF models were established by injecting mice with cyclophosphamide and busulfan. Then, ESCs-sEVs were intravenously transplanted into POF mice. The plasma of mice was harvested at 1 and 2 weeks after treatment to analyze the levels of anti-Mullerian hormone (AMH), estradiol (E2), and follicle stimulating hormone (FSH) by ELISA. The morphology of ovaries and follicles was observed by H&E staining, and apoptosis of granulosa cells was detected by TUNEL. In vitro, EdU and CCK-8 tests were used to evaluate the proliferation of cultured granulosa cells stimulated by ESCs-sEVs. Western blotting was used to determine the expression of PI3K/AKT and apoptotic-related proteins. RESULTS After transplantation of ESCs-sEVs, the levels of serum sex hormones recovered to normal levels. In addition, the number of follicles was significantly increased, and the number of apoptotic cells was decreased. The results in vitro revealed that ESCs-sEVs could significantly improve the proliferation rate of granulosa cells and increase the expression of phosphorylated PI3K and AKT. Meanwhile, the positive effect on proliferation and the negative effect on apoptosis observed in granulosa cells were obviously decreased when the PI3K/AKT signaling pathway was inhibited. CONCLUSION Our findings suggested that ESCs-sEVs could improve ovarian function by regulating the PI3K/AKT signaling pathway, which could provide a promising clinical therapy for POF.
Collapse
Affiliation(s)
- Mengyu Liu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
- Medical College of Soochow University, Suzhou, 215006, China
| | - Yu Qiu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Zhuowei Xue
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Ruoyu Wu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Jie Li
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Xin Niu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Ji Yuan
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China.
| | - Qingkai Wu
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No.600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
36
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|