251
|
Ng KS, Smith JA, McAteer MP, Mead BE, Ware J, Jackson FO, Carter A, Ferreira L, Bure K, Rowley JA, Reeve B, Brindley DA, Karp JM. Bioprocess decision support tool for scalable manufacture of extracellular vesicles. Biotechnol Bioeng 2019; 116:307-319. [PMID: 30063243 PMCID: PMC6322973 DOI: 10.1002/bit.26809] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/28/2022]
Abstract
Newly recognized as natural nanocarriers that deliver biological information between cells, extracellular vesicles (EVs), including exosomes and microvesicles, provide unprecedented therapeutic opportunities. Large-scale and cost-effective manufacturing is imperative for EV products to meet commercial and clinical demands; successful translation requires careful decisions that minimize financial and technological risks. Here, we develop a decision support tool (DST) that computes the most cost-effective technologies for manufacturing EVs at different scales, by examining the costs of goods associated with using published protocols. The DST identifies costs of labor and consumables during EV harvest as key cost drivers, substantiating a need for larger-scale, higher-throughput, and automated technologies for harvesting EVs. Importantly, we highlight a lack of appropriate technologies for meeting clinical demands, and propose a potentially cost-effective solution. This DST can facilitate decision-making very early on in development and be used to predict, and better manage, the risk of process changes when commercializing EV products.
Collapse
Affiliation(s)
- Kelvin S. Ng
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- RoosterBioFrederickMaryland
| | - James A. Smith
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Matthew P. McAteer
- Department of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusetts
| | - Benjamin E. Mead
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- Broad Institute of Harvard and MITCambridgeMassachusetts
- Koch Institute for Integrative Cancer Research, MITCambridgeMassachusetts
| | - Jamie Ware
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Felix O. Jackson
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | - Alison Carter
- Department of PaediatricsUniversity of OxfordOxfordUK
| | - Lino Ferreira
- University of Coimbra, Center for Neuroscience and Cell BiologyPortugal
| | - Kim Bure
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
| | | | - Brock Reeve
- Harvard Stem Cell InstituteCambridgeMassachusetts
| | - David A. Brindley
- Harvard Stem Cell InstituteCambridgeMassachusetts
- The Oxford‐UCL Centre for the Advancement of Sustainable Medical Innovation, University of OxfordOxfordUK
- Department of PaediatricsUniversity of OxfordOxfordUK
- Centre for Behavioural Medicine, UCL School of Pharmacy, University College LondonLondonUK
- UCSF‐Stanford Center of Excellence in Regulatory Science and InnovationSan FranciscoCalifornia
| | - Jeffrey M. Karp
- Harvard‐MIT Division of Health Sciences and TechnologyCambridgeMassachusetts
- Division of Engineering in Medicine, Department of MedicineBrigham & Women’s Hospital, Harvard Medical SchoolBostonMA
- Harvard Stem Cell InstituteCambridgeMassachusetts
- Broad Institute of Harvard and MITCambridgeMassachusetts
| |
Collapse
|
252
|
Narbute K, Piļipenko V, Pupure J, Dzirkale Z, Jonavičė U, Tunaitis V, Kriaučiūnaitė K, Jarmalavičiūtė A, Jansone B, Kluša V, Pivoriūnas A. Intranasal Administration of Extracellular Vesicles Derived from Human Teeth Stem Cells Improves Motor Symptoms and Normalizes Tyrosine Hydroxylase Expression in the Substantia Nigra and Striatum of the 6-Hydroxydopamine-Treated Rats. Stem Cells Transl Med 2019; 8:490-499. [PMID: 30706999 PMCID: PMC6477008 DOI: 10.1002/sctm.18-0162] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 12/19/2018] [Indexed: 01/09/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder affecting millions of people worldwide. At present, there is no effective cure for PD; treatments are symptomatic and do not halt progression of neurodegeneration. Extracellular vesicles (EVs) can cross the blood-brain barrier and represent promising alternative to the classical treatment strategies. In the present study, we examined therapeutic effects of intranasal administration of EVs derived from human exfoliated deciduous teeth stem cells (SHEDs) on unilateral 6-hydroxydopamine (6-OHDA) medial forebrain bundle (MFB) rat model of PD. CatWalk gait tests revealed that EVs effectively suppressed 6-OHDA-induced gait impairments. All tested gait parameters (stand, stride length, step cycle, and duty cycle) were significantly improved in EV-treated animals when compared with 6-OHDA-lesion group rats. Furthermore, EVs slowed down numbers of 6-OHDA-induced contralateral rotations in apomorphine test. Improvements in motor function correlated with normalization of tyrosine hydroxylase expression in the striatum and substantia nigra. In conclusion, we demonstrated, for the first time, the therapeutic efficacy of intranasal administration of EVs derived from SHEDs in a rat model of PD induced by 6-OHDA intra-MFB lesion. Our findings could be potentially exploited for the development of new treatment strategies against PD.
Collapse
Affiliation(s)
- Karīna Narbute
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Vladimirs Piļipenko
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Jolanta Pupure
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Zane Dzirkale
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Ugnė Jonavičė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Virginijus Tunaitis
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Karolina Kriaučiūnaitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Akvilė Jarmalavičiūtė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| | - Baiba Jansone
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Vija Kluša
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Riga, Latvia
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
| |
Collapse
|
253
|
Erkan EP, Saydam N, Chen CC, Saydam O. Extracellular Vesicles as Carriers of Suicide mRNA and/or Protein in Cancer Therapy. Methods Mol Biol 2019; 1895:87-96. [PMID: 30539531 DOI: 10.1007/978-1-4939-8922-5_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Gene therapy involves the introduction of genes (termed transgenes) into cells to compensate for a deficiency or to make a beneficial protein. Gene therapy can used as a form of cancer treatment. A particularly attractive paradigm in this regard involves the selective introduction of transgenes into cancer cells that converts inactive prodrugs into active chemotherapeutic agents, thereby triggering the death of cancer cells. Since prodrugs are inactive, they tend not to cause significant side-effects and are well-tolerated by patients relative to conventional chemotherapy. Several viral and nonviral vectors have been used as delivery tools for suicide gene therapy. Extracellular vesicles (EVs) are now recognized as a promising class of nonviral delivery vectors. Here, we describe a method in which a suicide fusion gene construct is loaded into EVs derived from a non-tumorigenic cell line. Delivery of these modified EVs to glioblastoma cell lines and spheroids decreases glioblastoma cell viability, induces apoptotic cell death, and inhibits tumor growth in vivo.
Collapse
Affiliation(s)
- Erdogan Pekcan Erkan
- Department of Medical Genetics, Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nurten Saydam
- Department of Neurosurgery and School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Clark C Chen
- Department of Neurosurgery and School of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Okay Saydam
- Department of Neurosurgery and School of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
254
|
Sayes CM, Hickey AJ. Perspectives for Characterizing Drug Component of Theranostic Products Containing Nanomaterials. Bioanalysis 2019. [DOI: 10.1007/978-3-030-01775-0_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
255
|
Pacienza N, Lee RH, Bae EH, Kim DK, Liu Q, Prockop DJ, Yannarelli G. In Vitro Macrophage Assay Predicts the In Vivo Anti-inflammatory Potential of Exosomes from Human Mesenchymal Stromal Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 13:67-76. [PMID: 30719485 PMCID: PMC6350420 DOI: 10.1016/j.omtm.2018.12.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023]
Abstract
Extracellular vesicles (EVs) play key roles in cell biology and may provide new clinical diagnostics and therapies. However, it has proven difficult to develop protocols for their purification and characterization. One of the major barriers in the field has been a lack of convenient assays for their bioactivity. Developing assays has not been a trivial matter, because of the heterogeneity of EVs, the multiple activities they demonstrate, and the uncertainty about their modes of action. Therefore, it is likely that multiple assays for their activities are needed. One important assay will be for the anti-inflammatory activity observed in mice after administration of the small EVs commonly referred to as exosomes. We developed an assay for the anti-inflammatory activity of exosomes with a line of mouse macrophages. The assay makes it possible to rank different preparations of exosomes by their anti-inflammatory activity, and their ranking predicts their efficacy in suppressing LPS-stimulated inflammation in mice. The assay is convenient for comparing multiple samples and, therefore, should be useful in developing protocols for the purification and characterization of anti-inflammatory exosomes.
Collapse
Affiliation(s)
- Natalia Pacienza
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA.,Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro/CONICET, Buenos Aires, Argentina
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA
| | - Eun-Hye Bae
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA
| | - Dong-Ki Kim
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA
| | - Qisong Liu
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA
| | - Darwin J Prockop
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA
| | - Gustavo Yannarelli
- Institute for Regenerative Medicine, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX 77845, USA.,Laboratorio de Regulación Génica y Células Madre, Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMeTTyB), Universidad Favaloro/CONICET, Buenos Aires, Argentina
| |
Collapse
|
256
|
Cheng Q, Shi X, Han M, Smbatyan G, Lenz HJ, Zhang Y. Reprogramming Exosomes as Nanoscale Controllers of Cellular Immunity. J Am Chem Soc 2018; 140:16413-16417. [PMID: 30452238 DOI: 10.1021/jacs.8b10047] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Exosomes are naturally occurring membranous vesicles secreted by various types of cells. Given their unique and important biological and pharmacological properties, exosomes have been emerging as a promising form of nanomedicine acting via efficient delivery of endogenous and exogenous therapeutics. Here we explore a new concept of utilizing endogenously derived exosomes as artificial controllers of cellular immunity to redirect and activate cytotoxic T cells toward cancer cells for killing. This was achieved through genetically displaying two distinct types of antibodies on exosomal surface. The resulting synthetic multivalent antibodies retargeted exosomes (SMART-Exos), which express monoclonal antibodies specific for T-cell CD3 and cancer cell-associated epidermal growth factor receptor (EGFR), were shown to not only induce cross-linking of T cells and EGFR-expressing breast cancer cells but also elicit potent antitumor immunity both in vitro and in vivo. This proof-of-concept study demonstrates a novel application of exosomes in cancer immunotherapy and may provide a general and versatile approach for the development of a new class of cell-free therapy.
Collapse
Affiliation(s)
- Qinqin Cheng
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States
| | - Xiaojing Shi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States
| | - Menglu Han
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States
| | - Goar Smbatyan
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine , University of Southern California , Los Angeles , California 90089 , United States
| | - Yong Zhang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy , University of Southern California , Los Angeles , California 90089 , United States.,Department of Chemistry, Dornsife College of Letters, Arts and Sciences , University of Southern California , Los Angeles , California 90089 , United States.,Norris Comprehensive Cancer Center , University of Southern California , Los Angeles , California 90089 , United States.,Research Center for Liver Diseases , University of Southern California , Los Angeles , California 90089 , United States
| |
Collapse
|
257
|
Fonsato V, De Lena M, Tritta S, Brossa A, Calvetti R, Tetta C, Camussi G, Bussolati B. Human liver stem cell-derived extracellular vesicles enhance cancer stem cell sensitivity to tyrosine kinase inhibitors through Akt/mTOR/PTEN combined modulation. Oncotarget 2018; 9:36151-36165. [PMID: 30546834 PMCID: PMC6281417 DOI: 10.18632/oncotarget.26319] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/24/2018] [Indexed: 01/18/2023] Open
Abstract
It is well recognized that Cancer Stem Cells (CSCs) sustain the initiation, the maintenance and the recurrence of tumors. We previously reported that extracellular vesicles (EVs) derived from human liver stem cells (HLSCs) were able to limit tumor development. In this study, we evaluated whether EV derived from HLSCs could act in synergy with tyrosine kinase inhibitors (TKIs) on apoptosis of CSCs isolated from renal carcinomas. For this purpose, we administered to renal CSCs, HLSC-EVs and TKIs, as co-incubation or sequential administration. We found that HLSC-EVs in combination with Sunitinb or Sorafenib significantly increased renal CSCs apoptosis induced by low TKI dose. At variance, no synergistic effect was observed when bone marrow mesenchymal stem cell-derived EVs were used. In particular, renal CSCs chemosensitivity to TKIs was enhanced when HLSC-EVs were either co-administered with TKIs or added after, but not before. CSC apoptosis was also incremented at a percentage comparable to that of co-administration when TKIs were loaded in HLSC-EVs. By a mechanistic point of view, Akt/mTOR and Erk and Creb intracellular pathways, known to be pivotal in the induction of tumor growth and survival, appeared modulated as consequence of TKIs/HLSC-EVs co-administration. Together, our results indicate that the synergistic effect of HLSC-EVs with TKIs may increase the response to TKIs at low doses, providing a rational for their combined use in the treatment of renal carcinoma.
Collapse
Affiliation(s)
- Valentina Fonsato
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Michela De Lena
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Stefania Tritta
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alessia Brossa
- 2i3T, Società per la gestione dell'incubatore di imprese e per il trasferimento tecnologico, Scarl University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Ruggero Calvetti
- Department of Molecular Biotechnology and Health Sciences, Torino, Italy
| | | | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, Torino, Italy
| | | |
Collapse
|
258
|
Gao D, Jiang L. Exosomes in cancer therapy: a novel experimental strategy. Am J Cancer Res 2018; 8:2165-2175. [PMID: 30555736 PMCID: PMC6291654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/16/2018] [Indexed: 06/09/2023] Open
Abstract
Exosomes are small membrane vesicles of endocytic origin secreted by most cell types. They play important roles in intercellular communications and many physiological processes. DCs-derived exosomes can prime naïve T cells and activate NK cells to shrink the tumor. Tumor-derived exosomes carry a variety of tumor antigens that trigger the robust tumor antigen-specific immune response. Tumor-derived exosomes also contain metastasis or invasive-related molecules, which maybe potential targets for cancer immunotherapy. Effector T cells-derived exosomes possess cytotoxic activity of their original cells, thus cause tumor cells lysis. In this review, we summarized the recent advances on the biogenesis and composition of exosomes, the functions of anti-tumor immune response, and the promising applications on cancer immunotherapy of exosomes from different origins. Exosomes schlep efficient targets homing to tumor sites and tend to be a promising new tool of immunotherapy to fight cancer in a cell-free system.
Collapse
Affiliation(s)
- Dong Gao
- Research Institute of Shenzhen Beike Biotechnology Co., Ltd.Keyuan Road 18, Shenzhen, Guangdong, P. R. China
- Shenzhen Hornetcorn Biotechnology Co., Ltd.Shihua Road 14, Shenzhen, Guangdong, P. R. China
| | - Lingling Jiang
- Shenzhen Hornetcorn Biotechnology Co., Ltd.Shihua Road 14, Shenzhen, Guangdong, P. R. China
- Sir Run Run Shaw Hospital, College of Medicine, Zhejiang UniversityQingchun East Road 3, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
259
|
Li Y, Zhu ZY, Huang TT, Zhou YX, Wang X, Yang LQ, Chen ZA, Yu WF, Li PY. The peripheral immune response after stroke-A double edge sword for blood-brain barrier integrity. CNS Neurosci Ther 2018; 24:1115-1128. [PMID: 30387323 PMCID: PMC6490160 DOI: 10.1111/cns.13081] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 02/07/2023] Open
Abstract
The blood‐brain barrier (BBB) is a highly regulated interface that separates the peripheral circulation and the brain. It plays a vital role in regulating the trafficking of solutes, fluid, and cells at the blood‐brain interface and maintaining the homeostasis of brain microenvironment for normal neuronal activity. Growing evidence has led to the realization that ischemic stroke elicits profound immune responses in the circulation and the activation of multiple subsets of immune cells, which in turn affect both the early disruption and the later repair of the BBB after stroke. Distinct phenotypes or subsets of peripheral immune cells along with diverse intracellular mechanisms contribute to the dynamic changes of BBB integrity after stroke. This review focuses on the interaction between the peripheral immune cells and the BBB after ischemic stroke. Understanding their reciprocal interaction may generate new directions for stroke research and may also drive the innovation of easy accessible immune modulatory treatment strategies targeting BBB in the pursuit of better stroke recovery.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zi-Yu Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ting-Ting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yu-Xi Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Zeng-Ai Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Pei-Ying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
260
|
Karnati HK, Garcia JH, Tweedie D, Becker RE, Kapogiannis D, Greig NH. Neuronal Enriched Extracellular Vesicle Proteins as Biomarkers for Traumatic Brain Injury. J Neurotrauma 2018; 36:975-987. [PMID: 30039737 DOI: 10.1089/neu.2018.5898] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of injury-related death throughout the world and lacks effective treatment. Surviving TBI patients often develop neuropsychiatric symptoms, and the molecular mechanisms underlying the neuronal damage and recovery following TBI are not well understood. Extracellular vesicles (EVs) are membranous nanoparticles that are divided into exosomes (originating in the endosomal/multi-vesicular body [MVB] system) and microvesicles (larger EVs produced through budding of the plasma membrane). Both types of EVs are generated by all cells and are secreted into the extracellular environment, and participate in cell-to-cell communication and protein and RNA delivery. EVs enriched for neuronal origin can be harvested from peripheral blood samples and their contents quantitatively examined as a window to follow potential changes occurring in brain. Recent studies suggest that the levels of exosomal proteins and microRNAs (miRNAs) may represent novel biomarkers to support the clinical diagnosis and potential response to treatment for neurological disorders. In this review, we focus on the biogenesis of EVs, their molecular composition, and recent advances in research of their contents as potential diagnostic tools for TBI.
Collapse
Affiliation(s)
- Hanuma Kumar Karnati
- 1 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Joseph H Garcia
- 1 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - David Tweedie
- 1 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Robert E Becker
- 1 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland.,2 Aristea Translational Medicine Corporation, Park City, Utah
| | - Dimitrios Kapogiannis
- 3 Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| | - Nigel H Greig
- 1 Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland
| |
Collapse
|
261
|
Emam SE, Ando H, Abu Lila AS, Kobayashi S, Shimizu T, Okuhira K, Ishima Y, Ishida T. Doxorubicin Expands in Vivo Secretion of Circulating Exosome in Mice. Biol Pharm Bull 2018; 41:1078-1083. [PMID: 29962402 DOI: 10.1248/bpb.b18-00202] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Modulation of tumor immunity is a known factor in the antitumor activity of many chemotherapeutic agents. Exosomes are extracellular nanometric vesicles that are released by almost all types of cells, which includes cancer cells. These vesicles play a crucial role in tumor immunity. Many in vitro studies have reproduced the aggressive secretion of exosomes following treatment with conventional anticancer drugs. Nevertheless, how chemotherapeutic agents including nanomedicines such as Doxil® affect the in vivo secretion of exosomes is yet to be elucidated. In this study, the effect of intravenous injection of either free doxorubicin (DXR) or liposomal DXR formulation (Doxil®) on exosome secretion was evaluated in BALB/c mice. Exosomes were isolated from serum by using an ExoQuick™ kit. Free DXR treatment markedly increased serum exosome levels in a post-injection time-dependent manner, while Doxil® treatment did not. Exosomal size distribution and marker protein expressions (CD9, CD63, and TSG101) were studied. The physical/biological characteristics of treatment-induced exosomes were comparable to those of control mice. Interestingly, splenectomy significantly suppressed the copious exosomal secretions induced by free DXR. Collectively, our results indicate that conventional anticancer agents induce the secretion of circulating exosomes, presumably via stimulating immune cells of the spleen. As far as we know, this study represents the first report indicating that conventional chemotherapeutics may induce exosome secretion which might, in turn, contribute partly to the antitumor effect of chemotherapeutic agents.
Collapse
Affiliation(s)
- Sherif E Emam
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Amr Selim Abu Lila
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University.,Department of Pharmaceutics, College of Pharmacy, Hail University
| | - Shinya Kobayashi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Taro Shimizu
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Keiichiro Okuhira
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Yu Ishima
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University
| |
Collapse
|
262
|
Silva AKA, Perretta S, Perrod G, Pidial L, Lindner V, Carn F, Lemieux S, Alloyeau D, Boucenna I, Menasché P, Dallemagne B, Gazeau F, Wilhelm C, Cellier C, Clément O, Rahmi G. Thermoresponsive Gel Embedded with Adipose Stem-Cell-Derived Extracellular Vesicles Promotes Esophageal Fistula Healing in a Thermo-Actuated Delivery Strategy. ACS NANO 2018; 12:9800-9814. [PMID: 30231208 DOI: 10.1021/acsnano.8b00117] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Extracellular vesicles (EVs) are increasingly envisioned as the next generation of biological pro-regenerative nanotherapeutic agents, as has already been demonstrated for heart, kidney, liver, and brain tissues; lung injury repair; and skin regeneration. Herein, we explore another potential EV therapeutic application, fistula healing, together with a local minimally invasive delivery strategy. Allogenic extracellular vesicles (EVs) from adipose tissue-derived stromal cells (ASCs) are administered in a porcine fistula model through a thermoresponsive Pluronic F-127 (PF-127) gel, injected locally at 4 °C and gelling at body temperature to retain EVs in the entire fistula tract. Complete fistula healing is reported to be 100% for the gel plus EVs group, 67% for the gel group, and 0% for the control, supporting the therapeutic use of Pluronic F-127 gel alone or combined with EVs. However, only the combination of gel and EVs results in a statistically significant (i) reduction of fibrosis, (ii) decline of inflammatory response, (iii) decrease in the density of myofibroblasts, and (iv) increase of angiogenesis. Overall, we demonstrate that ASC-EV delivery into a PF-127 gel represents a successful local minimally invasive strategy to induce a therapeutic effect in a swine fistula model. Our study presents prospects for EV administration strategies and for the management of post-operative fistulas.
Collapse
Affiliation(s)
- Amanda K A Silva
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Silvana Perretta
- Department of Digestive and Endocrine Surgery , Hôpital Civil de Strasbourg, Institut de Recherche contre les Cancers de l'Appareil Digestif (IRCAD) , 67091 , Strasbourg , France
- IHU, Minimally Invasive Hybrid Surgical Institute , 67091 , Strasbourg , France
| | - Guillaume Perrod
- Laboratoire Imagerie de l'Angiogénèse, Plateforme d'Imagerie du Petit Animal, PARCC, INSERM U970, Université Sorbonne Paris Cité (USPC) , Université Paris Descartes , 56 rue Leblanc , 75015 , Paris , France
| | - Laetitia Pidial
- Laboratoire Imagerie de l'Angiogénèse, Plateforme d'Imagerie du Petit Animal, PARCC, INSERM U970, Université Sorbonne Paris Cité (USPC) , Université Paris Descartes , 56 rue Leblanc , 75015 , Paris , France
| | - Véronique Lindner
- Department of Pathology , Hôpital Hautepierre , 1, Avenue Molière , 67098 Strasbourg , France
| | - Florent Carn
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Shony Lemieux
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Damien Alloyeau
- Laboratoire Matériaux et Phénomènes Quantiques (MPQ) , UMR 7162 CNRS/Université Paris - Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Imane Boucenna
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Philippe Menasché
- Department of Cardiovascular Surgery , Hôpital Européen Georges Pompidou; Paris Cardiovascular Research Center, INSERM U970, Université Paris Descartes , Paris , 75015 France
| | - Bernard Dallemagne
- Department of Digestive and Endocrine Surgery , Hôpital Civil de Strasbourg, Institut de Recherche contre les Cancers de l'Appareil Digestif (IRCAD) , 67091 , Strasbourg , France
| | - Florence Gazeau
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | - Claire Wilhelm
- Laboratoire Matières et Systèmes Complexes (MSC), UMR 7057 CNRS, Université Sorbonne Paris Cité (USPC) , Université Paris-Diderot , 10 rue Alice Domon et Léonie Duquet , 75205 Paris cedex 13 , France
| | | | - Olivier Clément
- Laboratoire Imagerie de l'Angiogénèse, Plateforme d'Imagerie du Petit Animal, PARCC, INSERM U970, Université Sorbonne Paris Cité (USPC) , Université Paris Descartes , 56 rue Leblanc , 75015 , Paris , France
| | - Gabriel Rahmi
- Laboratoire Imagerie de l'Angiogénèse, Plateforme d'Imagerie du Petit Animal, PARCC, INSERM U970, Université Sorbonne Paris Cité (USPC) , Université Paris Descartes , 56 rue Leblanc , 75015 , Paris , France
| |
Collapse
|
263
|
Tengattini S. Chromatographic Approaches for Purification and Analytical Characterization of Extracellular Vesicles: Recent Advancements. Chromatographia 2018. [DOI: 10.1007/s10337-018-3637-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
264
|
Lapitz A, Arbelaiz A, Olaizola P, Aranburu A, Bujanda L, Perugorria MJ, Banales JM. Extracellular Vesicles in Hepatobiliary Malignancies. Front Immunol 2018; 9:2270. [PMID: 30369925 PMCID: PMC6194158 DOI: 10.3389/fimmu.2018.02270] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 09/12/2018] [Indexed: 12/15/2022] Open
Abstract
Primary hepatobiliary malignancies include a heterogeneous group of cancers with dismal prognosis, among which hepatocellular carcinoma (HCC), cholangiocarcinoma (CCA), and hepatoblastoma (HB) stand out. These tumors mainly arise from the malignant transformation of hepatocytes, cholangiocytes (bile duct epithelial cells) or hepatoblasts (embryonic liver progenitor cells), respectively. Early diagnosis, prognosis prediction and effective therapies are still a utopia for these diseases. Extracellular vesicles (EVs) are small membrane-enclosed spheres secreted by cells and present in biological fluids. They contain multiple types of biomolecules, such as proteins, RNA, DNA, metabolites and lipids, which make them a potential source of biomarkers as well as regulators of human pathobiology. In this review, the role of EVs in the pathogenesis of hepatobiliary cancers and their potential usefulness as disease biomarkers are highlighted. Moreover, the therapeutic value of EV regulation is discussed and future directions on basic and clinical research are indicated.
Collapse
MESH Headings
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Extracellular Vesicles/genetics
- Extracellular Vesicles/immunology
- Extracellular Vesicles/metabolism
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- MicroRNAs/genetics
- MicroRNAs/immunology
- Models, Genetic
- Models, Immunological
Collapse
Affiliation(s)
- Ainhoa Lapitz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Ander Arbelaiz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Paula Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Aitziber Aranburu
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
| | - Maria J. Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- “Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas” (CIBERehd), Carlos III National Institute of Health, Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
265
|
Namba Y, Sogawa C, Okusha Y, Kawai H, Itagaki M, Ono K, Murakami J, Aoyama E, Ohyama K, Asaumi JI, Takigawa M, Okamoto K, Calderwood SK, Kozaki KI, Eguchi T. Depletion of Lipid Efflux Pump ABCG1 Triggers the Intracellular Accumulation of Extracellular Vesicles and Reduces Aggregation and Tumorigenesis of Metastatic Cancer Cells. Front Oncol 2018; 8:376. [PMID: 30364132 PMCID: PMC6191470 DOI: 10.3389/fonc.2018.00376] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/22/2018] [Indexed: 12/21/2022] Open
Abstract
The ATP-binding cassette transporter G1 (ABCG1) is a cholesterol lipid efflux pump whose role in tumor growth has been largely unknown. Our transcriptomics revealed that ABCG1 was powerfully expressed in rapidly metastatic, aggregative colon cancer cells, in all the ABC transporter family members. Coincidently, genetic amplification of ABCG1 is found in 10–35% of clinical samples of metastatic cancer cases. Expression of ABCG1 was further elevated in three-dimensional tumoroids (tumor organoids) within stemness-enhancing tumor milieu, whereas depletion of ABCG1 lowered cellular aggregation and tumoroid growth in vitro as well as hypoxia-inducible factor 1α in cancer cells around the central necrotic areas in tumors in vivo. Notably, depletion of ABCG1 triggered the intracellular accumulation of extracellular vesicles (EVs) and regression of tumoroids. Collectively, these data suggest that ABCG1 plays a crucial role in tumorigenesis in metastatic cancer and that depletion of ABCG1 triggers tumor regression with the accumulation of EVs and their derivatives and cargos, implicating a novel ABCG1-targeting therapeutic strategy by which redundant and toxic substances may be accumulated in tumors leading to their regression.
Collapse
Affiliation(s)
- Yuri Namba
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Oral and Maxillofacial Radiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Chiharu Sogawa
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuka Okusha
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hotaka Kawai
- Department of Oral Pathology and Medicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Itagaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kisho Ono
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun Murakami
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Oral Diagnosis and Dentomaxillofacial Radiology, Okayama University Hospital, Okayama, Japan
| | - Eriko Aoyama
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazumi Ohyama
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Jun-Ichi Asaumi
- Department of Oral and Maxillofacial Radiology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kuniaki Okamoto
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Stuart K Calderwood
- Division of Molecular and Cellular Biology, Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ken-Ichi Kozaki
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takanori Eguchi
- Department of Dental Pharmacology, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Advanced Research Center for Oral and Craniofacial Sciences, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
266
|
Jesus S, Soares E, Cruz MT, Borges O. Exosomes as adjuvants for the recombinant hepatitis B antigen: First report. Eur J Pharm Biopharm 2018; 133:1-11. [PMID: 30287267 DOI: 10.1016/j.ejpb.2018.09.029] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 08/10/2018] [Accepted: 09/28/2018] [Indexed: 01/11/2023]
Abstract
Over the past few years, exosomes, a class of extracellular vesicles (EVs), have emerged as key players for inter-cellular communication ultimately modulating the behavior of target cells with countless outcomes. Nevertheless, the potential role of exosomes as vaccine adjuvants remains largely unexplored. Herein, we hypothesized that exosomes derived from immune cells may have an immunostimulatory effect and could constitute a good target towards the development of new fine-tuned vaccine adjuvants. To accomplish this goal, exosomes isolated from lipopolysaccharide endotoxin (LPS)-stimulated human monocytic cell line (THP-1) were characterized and tested for their non-specific immunostimulatory activity when administered subcutaneously to healthy mice; additionally, exosomes' vaccine adjuvant ability was also disclosed after their inclusion in vaccine formulations. The results obtained suggested that the isolated exosomes evoked a pro-inflammatory profile in spleen cells of healthy mice through the induction of cytokines such as tumor necrosis factor alpha (TNF-α), chemokine (C-C motif) ligand 5 (CCL5, also known as RANTES) and interleukin 1 beta (IL-1β). Moreover, subcutaneous vaccination of mice with exosomes combined with a solution of hepatitis B recombinant antigen (HBsAg) or combined with a suspension containing HBsAg loaded poly-ε-caprolactone (PCL)/chitosan nanoparticles (NPs), induced a humoral immune response quite similar to the one achieved with the experimental control group (HBsAg solution without exosomes). However, exosomes triggered an immunomodulator effect on the cellular immune response, highlighted by the enhancement of IFN-γ secretion. To the best of authors knowledge, this is the first report describing extensively the role of unmodified exosomes as adjuvants and co-adjuvants for hepatitis B vaccination strategies.
Collapse
Affiliation(s)
- Sandra Jesus
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Edna Soares
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, Faculty of Medicine, 1st floor, 3004-504 Coimbra, Portugal.
| |
Collapse
|
267
|
Zhang S, Dai H, Zhu L, Lin F, Hu Z, Jing R, Zhang W, Zhao C, Hong X, Zhong JH, Pan L. Microvesicles packaging IL-1β and TNF-α enhance lung inflammatory response to mechanical ventilation in part by induction of cofilin signaling. Int Immunopharmacol 2018; 63:74-83. [PMID: 30075431 DOI: 10.1016/j.intimp.2018.07.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/13/2018] [Accepted: 07/26/2018] [Indexed: 12/18/2022]
Abstract
Microvesicles shed from pulmonary cells are capable of transferring inflammatory cargo to recipient cells nearby or in distant to enhance inflammation. Some authors believe that cofilin controls actin dynamics and regulates vesicle mobilization. We therefore investigated the potential role and mechanism of microvesicles in ventilator-induced lung injury (VILI). Fifty male C57BL/6 mice were orotracheally intubated and either allowed to breathe spontaneously or they were mechanically ventilated with different tidal volumes (Vt) and ventilation times. Lung tissue injury was assessed in terms of lung histopathologic examination, wet/dry weight ratios, and levels of total proteins and of cytokines. Microvesicle characteristics, sizes, contents and levels as well as cofilin were also measured. We found that lung inflammation increased significantly after ventilation with high Vt for 4 h; these conditions led to secretion of larger and more microvesicles into the alveoli than animals with/without ventilation at low Vt. Intratracheal instillation of microvesicles obtained from animals ventilated with low or high Vt triggered significant lung inflammation in naive mice, and these high-Vt microvesicles not only carried more IL-1β and TNF-α but also induced more severe lung inflammation compared to low-Vt microvesicles; And high-Vt microvesicles at 2 h carried more molecular cargo than that at 1 h or 4 h, which may involve the shift and amplification of inflammation. Furthermore, blocking the phosphorylation of cofilin can not only inhibit microvesicle formation in the lung, but also reduce lung injury. Collectively, our data suggest that microvesicles packaging IL-1β and TNF-α enhance lung inflammation in VILI.
Collapse
Affiliation(s)
- Suisui Zhang
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Huijun Dai
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Lingyu Zhu
- Department of Breast Surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Fei Lin
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhaokun Hu
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Ren Jing
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Weikang Zhang
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chen Zhao
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xueqi Hong
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Hong Zhong
- Department of Hepatobiliary Surgery, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China
| | - Linghui Pan
- Department of Anesthesiology, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
268
|
Could hypoxia influence basic biological properties and ultrastructural features of adult canine mesenchymal stem /stromal cells? Vet Res Commun 2018; 42:297-308. [PMID: 30238341 DOI: 10.1007/s11259-018-9738-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
The aim of the present study was to compare canine adipose tissue mesenchymal stem cells cultured under normoxic (20% O2) and not severe hypoxic (7% O2) conditions in terms of marker expression, proliferation rate, differentiation potential and cell morphology. Intra-abdominal fat tissue samples were recovered from 4 dogs and cells isolated from each sample were cultured under hypoxic and normoxic conditions. Proliferation rate and adhesion ability were determined, differentiation towards chondrogenic, osteogenic and adipogenic lineages was induced; the expression of CD44, CD34, DLA-DQA1, DLA-DRA1 was determined by PCR, while flow cytometry analysis for CD90, CD105, CD45 and CD14 was carried out. The morphological study was performed by transmission electron microscopy. Canine AT-MSCs, cultured under different oxygen tensions, maintained their basic biological features. However, under hypoxia, cells were not able to form spheroid aggregates revealing a reduction of their adhesivness. In both conditions, MSCs mainly displayed the same ultrastructural morphology and retained the ability to produce membrane vesicles. Noteworthy, MSCs cultivated under hypoxya revealed a huge shedding of large complex vesicles, containing smaller round-shaped vesicles. In our study, hypoxia partially influences the basic biological properties and the ultrastructural features of canine mesenchymal stem /stromal cells. Further studies are needed to clarify how hypoxia affects EVs production in term of amount and content in order to understand its contribution in tissue regenerative mechanisms and the possible employment in clinical applications. The findings of the present work could be noteworthy for canine as well as for other mammalian species.
Collapse
|
269
|
Buzás EI, Tóth EÁ, Sódar BW, Szabó-Taylor KÉ. Molecular interactions at the surface of extracellular vesicles. Semin Immunopathol 2018; 40:453-464. [PMID: 29663027 PMCID: PMC6208672 DOI: 10.1007/s00281-018-0682-0] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/26/2018] [Indexed: 12/21/2022]
Abstract
Extracellular vesicles such as exosomes, microvesicles, apoptotic bodies, and large oncosomes have been shown to participate in a wide variety of biological processes and are currently under intense investigation in many different fields of biomedicine. One of the key features of extracellular vesicles is that they have relatively large surface compared to their volume. Some extracellular vesicle surface molecules are shared with those of the plasma membrane of the releasing cell, while other molecules are characteristic for extracellular vesicular surfaces. Besides proteins, lipids, glycans, and nucleic acids are also players of extracellular vesicle surface interactions. Being secreted and present in high number in biological samples, collectively extracellular vesicles represent a uniquely large interactive surface area which can establish contacts both with cells and with molecules in the extracellular microenvironment. Here, we provide a brief overview of known components of the extracellular vesicle surface interactome and highlight some already established roles of the extracellular vesicle surface interactions in different biological processes in health and disease.
Collapse
Affiliation(s)
- Edit I Buzás
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary.
- MTA-SE Immune-Proteogenomics Research Group, Budapest, Hungary.
| | - Eszter Á Tóth
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Barbara W Sódar
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Katalin É Szabó-Taylor
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
270
|
Miura N, Ueda M. Evaluation of Unconventional Protein Secretion by Saccharomyces cerevisiae and other Fungi. Cells 2018; 7:cells7090128. [PMID: 30200367 PMCID: PMC6162777 DOI: 10.3390/cells7090128] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 12/11/2022] Open
Abstract
Development of proteome analysis of extracellular proteins has revealed that a wide variety of proteins, including fungal allergens are present outside the cell. These secreted allergens often do not contain known secretion signal sequences. Recent research progress shows that some fungal allergens are secreted by unconventional secretion pathways, including autophagy- and extracellular-vesicle-dependent pathways. However, secretion pathways remain unknown for the majority of extracellular proteins. This review summarizes recent data on unconventional protein secretion in Saccharomyces cerevisiae and other fungi. Particularly, methods for evaluating unconventional protein secretion are proposed for fungal species, including S. cerevisiae, a popular model organism for investigating protein secretion pathways.
Collapse
Affiliation(s)
- Natsuko Miura
- Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai 599-8531, Japan.
| | - Mitsuyoshi Ueda
- Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
271
|
Wang L, Kempton JB, Brigande JV. Gene Therapy in Mouse Models of Deafness and Balance Dysfunction. Front Mol Neurosci 2018; 11:300. [PMID: 30210291 PMCID: PMC6123355 DOI: 10.3389/fnmol.2018.00300] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022] Open
Abstract
Therapeutic strategies to restore hearing and balance in mouse models of inner ear disease aim to rescue sensory function by gene replacement, augmentation, knock down or knock out. Modalities to achieve therapeutic effects have utilized virus-mediated transfer of wild type genes and small interfering ribonucleic acids; systemic and focal administration of antisense oligonucleotides (ASO) and designer small molecules; and lipid-mediated transfer of Cas 9 ribonucleoprotein (RNP) complexes. This work has established that gene or drug administration to the structurally and functionally immature, early neonatal mouse inner ear prior to hearing onset is a prerequisite for the most robust therapeutic responses. These observations may have significant implications for translating mouse inner ear gene therapies to patients. The human fetus hears by gestational week 19, suggesting that a corollary window of therapeutic efficacy closes early in the second trimester of pregnancy. We hypothesize that fetal therapeutics deployed prior to hearing onset may be the most effective approach to preemptively manage genetic mutations that cause deafness and vestibular dysfunction. We assert that gene therapy studies in higher vertebrate model systems with fetal hearing onset and a comparable acoustic range and sensitivity to that of humans are an essential step to safely and effectively translate murine gene therapies to the clinic.
Collapse
Affiliation(s)
- Lingyan Wang
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| | - J Beth Kempton
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| | - John V Brigande
- Oregon Hearing Research Center, Department of Otolaryngology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
272
|
Pick H, Alves AC, Vogel H. Single-Vesicle Assays Using Liposomes and Cell-Derived Vesicles: From Modeling Complex Membrane Processes to Synthetic Biology and Biomedical Applications. Chem Rev 2018; 118:8598-8654. [PMID: 30153012 DOI: 10.1021/acs.chemrev.7b00777] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The plasma membrane is of central importance for defining the closed volume of cells in contradistinction to the extracellular environment. The plasma membrane not only serves as a boundary, but it also mediates the exchange of physical and chemical information between the cell and its environment in order to maintain intra- and intercellular functions. Artificial lipid- and cell-derived membrane vesicles have been used as closed-volume containers, representing the simplest cell model systems to study transmembrane processes and intracellular biochemistry. Classical examples are studies of membrane translocation processes in plasma membrane vesicles and proteoliposomes mediated by transport proteins and ion channels. Liposomes and native membrane vesicles are widely used as model membranes for investigating the binding and bilayer insertion of proteins, the structure and function of membrane proteins, the intramembrane composition and distribution of lipids and proteins, and the intermembrane interactions during exo- and endocytosis. In addition, natural cell-released microvesicles have gained importance for early detection of diseases and for their use as nanoreactors and minimal protocells. Yet, in most studies, ensembles of vesicles have been employed. More recently, new micro- and nanotechnological tools as well as novel developments in both optical and electron microscopy have allowed the isolation and investigation of individual (sub)micrometer-sized vesicles. Such single-vesicle experiments have revealed large heterogeneities in the structure and function of membrane components of single vesicles, which were hidden in ensemble studies. These results have opened enormous possibilities for bioanalysis and biotechnological applications involving unprecedented miniaturization at the nanometer and attoliter range. This review will cover important developments toward single-vesicle analysis and the central discoveries made in this exciting field of research.
Collapse
Affiliation(s)
- Horst Pick
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| | - Ana Catarina Alves
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| | - Horst Vogel
- Institute of Chemical Sciences and Engineering , Ecole Polytechnique Fédérale de Lausanne (EPFL) , CH-1015 Lausanne , Switzerland
| |
Collapse
|
273
|
Panagiotou N, Neytchev O, Selman C, Shiels PG. Extracellular Vesicles, Ageing, and Therapeutic Interventions. Cells 2018; 7:cells7080110. [PMID: 30126173 PMCID: PMC6115766 DOI: 10.3390/cells7080110] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023] Open
Abstract
A more comprehensive understanding of the human ageing process is required to help mitigate the increasing burden of age-related morbidities in a rapidly growing global demographic of elderly individuals. One exciting novel strategy that has emerged to intervene involves the use of extracellular vesicles to engender tissue regeneration. Specifically, this employs their molecular payloads to confer changes in the epigenetic landscape of ageing cells and ameliorate the loss of functional capacity. Understanding the biology of extracellular vesicles and the specific roles they play during normative ageing will allow for the development of novel cell-free therapeutic interventions. Hence, the purpose of this review is to summarise the current understanding of the mechanisms that drive ageing, critically explore how extracellular vesicles affect ageing processes and discuss their therapeutic potential to mitigate the effects of age-associated morbidities and improve the human health span.
Collapse
Affiliation(s)
- Nikolaos Panagiotou
- Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - Ognian Neytchev
- Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| | - Colin Selman
- College of Medical, Veterinary & Life Sciences, Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Graham Kerr, Glasgow G12 8QQ, UK.
| | - Paul G Shiels
- Wolfson Wohl Cancer Research Centre, College of Medical, Veterinary & Life Sciences, Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
274
|
Frank J, Richter M, de Rossi C, Lehr CM, Fuhrmann K, Fuhrmann G. Extracellular vesicles protect glucuronidase model enzymes during freeze-drying. Sci Rep 2018; 8:12377. [PMID: 30120298 PMCID: PMC6098026 DOI: 10.1038/s41598-018-30786-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 08/02/2018] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are natural nanoparticles that play important roles in intercellular communication and are increasingly studied for biosignalling, pathogenesis and therapy. Nevertheless, little is known about optimal conditions for their transfer and storage, and the potential impact on preserving EV-loaded cargoes. We present the first comprehensive stability assessment of different widely available types of EVs during various storage conditions including -80 °C, 4 °C, room temperature, and freeze-drying (lyophilisation). Lyophilisation of EVs would allow easy handling at room temperature and thus significantly enhance their expanded investigation. A model enzyme, β-glucuronidase, was loaded into different types of EVs derived from mesenchymal stem cells, endothelial cells and cancer cells. Using asymmetric flow field-flow fractionation we proved that the model enzyme is indeed stably encapsulated into EVs. When assessing enzyme activity as indicator for EV stability, and in comparison to liposomes, we show that EVs are intrinsically stable during lyophilisation, an effect further enhanced by cryoprotectants. Our findings provide new insight for exploring lyophilisation as a novel storage modality and we create an important basis for standardised and advanced EV applications in biomedical research.
Collapse
Affiliation(s)
- Julia Frank
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123, Saarbrücken, Germany
| | - Maximilian Richter
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Chiara de Rossi
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Drug Delivery (DDEL), Campus E8.1, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Department of Drug Delivery (DDEL), Campus E8.1, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123, Saarbrücken, Germany
| | - Kathrin Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123, Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123, Saarbrücken, Germany.
| |
Collapse
|
275
|
Kosgodage US, Mould R, Henley AB, Nunn AV, Guy GW, Thomas EL, Inal JM, Bell JD, Lange S. Cannabidiol (CBD) Is a Novel Inhibitor for Exosome and Microvesicle (EMV) Release in Cancer. Front Pharmacol 2018; 9:889. [PMID: 30150937 PMCID: PMC6099119 DOI: 10.3389/fphar.2018.00889] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 07/23/2018] [Indexed: 01/05/2023] Open
Abstract
Exosomes and microvesicles (EMV) are lipid bilayer-enclosed structures, released by cells and involved in intercellular communication through transfer of proteins and genetic material. EMV release is also associated with various pathologies, including cancer, where increased EMV release is amongst other associated with chemo-resistance and active transfer of pro-oncogenic factors. Recent studies show that EMV-inhibiting agents can sensitize cancer cells to chemotherapeutic agents and reduce cancer growth in vivo. Cannabidiol (CBD), a phytocannabinoid derived from Cannabis sativa, has anti-inflammatory and anti-oxidant properties, and displays anti-proliferative activity. Here we report a novel role for CBD as a potent inhibitor of EMV release from three cancer cell lines: prostate cancer (PC3), hepatocellular carcinoma (HEPG2) and breast adenocarcinoma (MDA-MB-231). CBD significantly reduced exosome release in all three cancer cell lines, and also significantly, albeit more variably, inhibited microvesicle release. The EMV modulating effects of CBD were found to be dose dependent (1 and 5 μM) and cancer cell type specific. Moreover, we provide evidence that this may be associated with changes in mitochondrial function, including modulation of STAT3 and prohibitin expression, and that CBD can be used to sensitize cancer cells to chemotherapy. We suggest that the known anti-cancer effects of CBD may partly be due to the regulatory effects on EMV biogenesis, and thus CBD poses as a novel and safe modulator of EMV-mediated pathological events.
Collapse
Affiliation(s)
- Uchini S Kosgodage
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, London, United Kingdom
| | - Rhys Mould
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Aine B Henley
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Alistair V Nunn
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Geoffrey W Guy
- GW Research, Sovereign House Vision Park, Cambridge, United Kingdom
| | - E L Thomas
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Jameel M Inal
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Jimmy D Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, Department of Biomedical Sciences, University of Westminster, London, United Kingdom.,Department of Pharmacology, University College London School of Pharmacy, London, United Kingdom
| |
Collapse
|
276
|
Familari M, Cronqvist T, Masoumi Z, Hansson SR. Placenta-derived extracellular vesicles: their cargo and possible functions. Reprod Fertil Dev 2018; 29:433-447. [PMID: 26411402 DOI: 10.1071/rd15143] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 08/29/2015] [Indexed: 12/15/2022] Open
Abstract
The literature on extracellular vesicles consists of rapidly expanding and often contradictory information. In this paper we attempt to review what is currently known regarding extracellular vesicles released specifically from human placental syncytiotrophoblast cells with a focus on the common but complex pregnancy-associated syndrome pre-eclampsia, where the level of syncytiotrophoblast extracellular vesicle release is significantly increased. We review common methods for syncytiotrophoblast extracellular vesicle derivation and isolation and we discuss the cargo of syncytiotrophoblast extracellular vesicles including proteins, RNA and lipids and their possible functions. A meta-analysis of available trophoblast-derived extracellular vesicle proteomic datasets revealed only three proteins in common: albumin, fibronectin-1 and plasminogen activator inhibitor-1, suggesting some variability in vesicle cargo, most likely reflecting stage and cell type of origin. We discuss the possible sources of variability that may have led to the low number of common markers, which has led us to speculate that markers and density in common use may not be strict criteria for identifying and isolating placenta-derived exosomes.
Collapse
Affiliation(s)
- Mary Familari
- School of Biosciences, University of Melbourne, Parkville, Vic. 3010, Australia
| | - Tina Cronqvist
- Lund University, Department of Clinical Sciences, Lund, Obstetrics and Gynecology, Klinikgatan 28, 221 85 Lund, Sweden
| | - Zahra Masoumi
- Lund University, Department of Clinical Sciences, Lund, Obstetrics and Gynecology, Klinikgatan 28, 221 85 Lund, Sweden
| | - Stefan R Hansson
- Lund University, Department of Clinical Sciences, Lund, Obstetrics and Gynecology, Klinikgatan 28, 221 85 Lund, Sweden
| |
Collapse
|
277
|
Rosso L, Zanella A, Righi I, Barilani M, Lazzari L, Scotti E, Gori F, Mendogni P. Lung transplantation, ex-vivo reconditioning and regeneration: state of the art and perspectives. J Thorac Dis 2018; 10:S2423-S2430. [PMID: 30123580 DOI: 10.21037/jtd.2018.04.151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Lung transplantation is the only therapeutic option for end-stage pulmonary failure. Nevertheless, the shortage of donor pool available for transplantation does not allow to satisfy the requests, thus the mortality on the waiting list remains high. One of the tools to overcome the donor pool shortage is the use of ex-vivo lung perfusion (EVLP) to preserve, evaluate and recondition selected lung grafts not otherwise suitable for transplantation. EVLP is nowadays a clinical reality and have several destinations of use. After a narrative review of the literature and looking at our experience we can assume that one of the chances to improve the outcome of lung transplantation and to overcome the donor pool shortage could be the tissue regeneration of the graft during EVLP and the immunomodulation of the recipient. Both these strategies are performed using mesenchymal stem cells (MSC). The results of the models of lung perfusion with MSC-based cell therapy open the way to a new innovative approach that further increases the potential for using of the lung perfusion platform.
Collapse
Affiliation(s)
- Lorenzo Rosso
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Alberto Zanella
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.,Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Righi
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Mario Barilani
- Unit of Regenerative Medicine-Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,EPIGET LAB, Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Lorenza Lazzari
- Unit of Regenerative Medicine-Cell Factory, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Eleonora Scotti
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Francesca Gori
- Department of Anesthesiology, Intensive Care and Emergency, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Mendogni
- Thoracic Surgery and Lung Transplant Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
278
|
Nooshabadi VT, Mardpour S, Yousefi-Ahmadipour A, Allahverdi A, Izadpanah M, Daneshimehr F, Ai J, Banafshe HR, Ebrahimi-Barough S. The extracellular vesicles-derived from mesenchymal stromal cells: A new therapeutic option in regenerative medicine. J Cell Biochem 2018; 119:8048-8073. [PMID: 29377241 DOI: 10.1002/jcb.26726] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are adult multipotent cells that due to their ability to homing to damaged tissues and differentiate into specialized cells, are remarkable cells in the field of regenerative medicine. It's suggested that the predominant mechanism of MSCs in tissue repair might be related to their paracrine activity. The utilization of MSCs for tissue repair is initially based on the differentiation ability of these cells; however now it has been revealed that only a small fraction of the transplanted MSCs actually fuse and survive in host tissues. Indeed, MSCs supply the microenvironment with the secretion of soluble trophic factors, survival signals and the release of extracellular vesicles (EVs) such as exosome. Also, the paracrine activity of EVs could mediate the cellular communication to induce cell-differentiation/self-renewal. Recent findings suggest that EVs released by MSCs may also be critical in the physiological function of these cells. This review provides an overview of MSC-derived extracellular vesicles as a hopeful opportunity to advance novel cell-free therapy strategies that might prevail over the obstacles and risks associated with the use of native or engineered stem cells. EVs are very stable; they can pass the biological barriers without rejection and can shuttle bioactive molecules from one cell to another, causing the exchange of genetic information and reprogramming of the recipient cells. Moreover, extracellular vesicles may provide therapeutic cargo for a wide range of diseases and cancer therapy.
Collapse
Affiliation(s)
| | - Soura Mardpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Yousefi-Ahmadipour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Allahverdi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Izadpanah
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Daneshimehr
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid R Banafshe
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
279
|
Giulietti M, Santoni M, Cimadamore A, Carrozza F, Piva F, Cheng L, Lopez-Beltran A, Scarpelli M, Battelli N, Montironi R. Exploring Small Extracellular Vesicles for Precision Medicine in Prostate Cancer. Front Oncol 2018; 8:221. [PMID: 29951374 PMCID: PMC6008382 DOI: 10.3389/fonc.2018.00221] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/29/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor microenvironment constitutes a complex network in which tumor cells communicate among them and with stromal and immune cells. It has been shown that cancer cells are able to exchange genetic materials through small extracellular vesicles (EVs), a heterogeneous group of vesicles with different size and shape, cargo content, and function. The importance to investigate populations of circulating EVs would be of great importance as prostate cancer (PCa) biomarkers. In several neoplasms as well as in PCa, nanometer-sized EVs of endosomal origin are implicated in supporting tumor growth and metastatic spread by both altering local stroma cells and creating a protumor environment that favors the formation of pre-metastatic niches. Several techniques are applicable for the isolation and analysis of PCa-derived small EVs and are illustrated in this article. Due to the high sensitivity and specificity of these techniques, small EVs have become ideal candidates for early diagnosis. Moreover, we discuss the role of small EVs during PCa carcinogenesis, as well as in modulating the development of drug resistance to hormonal therapy and chemotherapy, thus underlining the potential of EV-tailored strategies in PCa patients.
Collapse
Affiliation(s)
- Matteo Giulietti
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
280
|
Goes A, Fuhrmann G. Biogenic and Biomimetic Carriers as Versatile Transporters To Treat Infections. ACS Infect Dis 2018; 4:881-892. [PMID: 29553240 DOI: 10.1021/acsinfecdis.8b00030] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biogenic and biomimetic therapeutics are a relatively new class of systems that are of physiological origin and/or take advantage of natural pathways or aim at mimicking these to improve selective interaction with target tissue. The number of biogenic and bioengineered avenues for drug therapy and diagnostics has multiplied over the past years for many applications, indicating the high expectations associated with this biological route. Nevertheless, the use of "bio"-related approaches for treating or diagnosing infectious diseases is still rare. Given that infectious diseases, in particular bacterial resistances, are seriously on the rise, there is an urgent need to take advantage of biogenic and bioengineered systems to target these challenges. In this manuscript, we first give a definition of the various "bio" terms, including biogenic, biomimetic, bioinspired, and bioengineered and we highlight them using tangible applications in the field of infectious diseases. Our examples cover cell-derived systems, including bioengineered bacteria, virus-like particles, and different cell-mimetics. Moreover, we discuss natural and bioengineered particles such as extracellular vesicles from mammalian and bacterial sources and liposomes. A concluding section outlines the potential for biomaterial-related avenues to overcome challenges associated with difficult-to-treat infections. We critically discuss benefits and risks for these applications and give an outlook on the future of biogenic engineering.
Collapse
Affiliation(s)
- Adriely Goes
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI), Biogenic Nanotherapeutics group (BION), Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
| |
Collapse
|
281
|
Ottaviani L, Sansonetti M, da Costa Martins PA. Myocardial cell-to-cell communication via microRNAs. Noncoding RNA Res 2018; 3:144-153. [PMID: 30175287 PMCID: PMC6114265 DOI: 10.1016/j.ncrna.2018.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/30/2018] [Accepted: 05/25/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Lara Ottaviani
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Marida Sansonetti
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.,Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
282
|
Abstract
Biotechnology has revolutionized therapeutics for the treatment of a wide range of diseases. Recent advances in protein engineering and material science have made the targeted delivery of enzyme therapeutics using nanocarriers (NCs) a new model of treatment. Several NCs have been approved for clinical use in drug delivery. Despite their advantages, few NCs have been approved to deliver enzyme cargo in a targeted manner. This review details the current arsenal of platforms developed to deliver enzyme therapeutics as well as the advantages and challenges of using enzymes as drugs, with examples from the literature, and discusses the benefits and liabilities of a given approach. We conclude by providing a perspective on how this field may evolve over the near and long-term.
Collapse
|
283
|
Abstract
With the advancements in antenatal steroid therapies and surfactant replacement, current clinical practices in neonatal intensive care units allow the survival of infants at very low gestational age. Despite these advances, there continues to be significant morbidity associated with extreme preterm birth that includes both short-term and long-term cardiorespiratory impairment. With no effective single therapy in preventing or treating developmental lung injuries, the need for new tools to treat and reduce risk of complications associated with extreme preterm birth is urgent. Stem cell-based therapies, in particular therapies utilizing mesenchymal stem (stromal) cells (MSCs), have shown promise in a number of animal models of lung pathologies relevant to neonatology. Recent studies in this field have consolidated the concept that the therapeutic mechanism of MSC action is paracrine, and this led to wide acceptance of the concept that the delivery of the MSC secretome rather than live cells may provide an alternative therapeutic approach for many complex diseases. Here, we summarize the significance and application of cell-free based therapies in preclinical models of neonatal lung injury. We emphasize the development of extracellular vesicle (EV)-based therapeutics and focus on the challenges that remain to be addressed before their application to clinical practice.
Collapse
|
284
|
Differential Interaction of Platelet-Derived Extracellular Vesicles with Leukocyte Subsets in Human Whole Blood. Sci Rep 2018; 8:6598. [PMID: 29700367 PMCID: PMC5920058 DOI: 10.1038/s41598-018-25047-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/05/2018] [Indexed: 12/18/2022] Open
Abstract
Secretion and exchange of biomolecules via extracellular vesicles (EVs) are crucial mechanisms in intercellular communication, and the roles of EVs in infection, inflammation, or thrombosis have been increasingly recognized. EVs have emerged as central players in immune regulation and can enhance or suppress the immune response, depending on the state of donor and recipient cells. We investigated the interaction of blood cell-derived EVs with leukocyte subpopulations (monocytes and their subsets, granulocytes, B cells, T cells, and NK cells) directly in whole blood using a combination of flow cytometry, imaging flow cytometry, cell sorting, and high resolution confocal microscopy. Platelet-derived EVs constituted the majority of circulating EVs and were preferentially associated with granulocytes and monocytes, while they scarcely interacted with lymphocytes. Further flow cytometric differentiation of monocyte subsets provided clear indications for a preferential association of platelet-derived EVs with intermediate (CD14++CD16+) monocytes in whole blood.
Collapse
|
285
|
Chang YH, Wu KC, Harn HJ, Lin SZ, Ding DC. Exosomes and Stem Cells in Degenerative Disease Diagnosis and Therapy. Cell Transplant 2018; 27:349-363. [PMID: 29692195 PMCID: PMC6038041 DOI: 10.1177/0963689717723636] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Stroke can cause death and disability, resulting in a huge burden on society. Parkinson’s disease (PD) is a chronic neurodegenerative disorder characterized by motor dysfunction. Osteoarthritis (OA) is a progressive degenerative joint disease characterized by cartilage destruction and osteophyte formation in the joints. Stem cell therapy may provide a biological treatment alternative to traditional pharmacological therapy. Mesenchymal stem cells (MSCs) are preferred because of their differentiation ability and possible derivation from many adult tissues. In addition, the paracrine effects of MSCs play crucial anti-inflammatory and immunosuppressive roles in immune cells. Extracellular vesicles (EVs) are vital mediators of cell-to-cell communication. Exosomes contain various molecules such as microRNA (miRNA), which mediates biological functions through gene regulation. Therefore, exosomes carrying miRNA or other molecules can enhance the therapeutic effects of MSC transplantation. MSC-derived exosomes have been investigated in various animal models representing stroke, PD, and OA. Exosomes are a subtype of EVs. This review article focuses on the mechanism and therapeutic potential of MSC-derived exosomes in stroke, PD, and OA in basic and clinical aspects.
Collapse
Affiliation(s)
- Yu-Hsun Chang
- 1 Department of Pediatrics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan.,2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Kung-Chi Wu
- 3 Department of Orthopedics, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Horng-Jyh Harn
- 4 Department of Pathology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Shinn-Zong Lin
- 5 Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Dah-Ching Ding
- 2 Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.,6 Department of Obstetrics and Gynecology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| |
Collapse
|
286
|
Li L, Sun Y, Feng M, Wang L, Liu J. Clinical significance of blood-based miRNAs as biomarkers of non-small cell lung cancer. Oncol Lett 2018; 15:8915-8925. [PMID: 29805626 DOI: 10.3892/ol.2018.8469] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 01/05/2018] [Indexed: 12/18/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) accounts for 85% of all cases of lung cancer. However, the predicted 5-year survival rate of patients with NSCLC is only 15.9%. microRNAs (miRNAs) are single-stranded, noncoding RNA molecules that are easily detectable in blood in a non-invasive manner, with features of stability, reproducibility and consistency in blood. Therefore, miRNAs derived from blood are able to have a significant impact on NSCLC diagnosis, metastasis and targeted therapies. Compared with the clinical protein markers carcinoembryonic antigen, cytokeratin fragment 21-1 and cancer antigen-125, blood-based miRNAs also display a higher diagnostic efficacy in NSCLC. Exosomal miRNAs are identified to be easily measured and have the potential to be used as diagnostic biomarkers in NSCLC, therefore providing an alternative method of biopsy profiling. The miRNA profile in exosomes is similar to the profile in primary tumor, meaning that this feature may be a powerful tool for NSCLC clinical diagnosis and targeted therapies. The focus of the present review was the clinical significance of blood-based exosomal miRNAs in diagnosis, prognosis, metastasis and targeted therapies of NSCLC.
Collapse
Affiliation(s)
- Lin Li
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Centre, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Yu Sun
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Centre, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Min Feng
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Centre, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Liang Wang
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Centre, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jing Liu
- Stem Cell Clinical Research Center, National Joint Engineering Laboratory, Regenerative Medicine Centre, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| |
Collapse
|
287
|
Webb RL, Kaiser EE, Jurgielewicz BJ, Spellicy S, Scoville SL, Thompson TA, Swetenburg RL, Hess DC, West FD, Stice SL. Human Neural Stem Cell Extracellular Vesicles Improve Recovery in a Porcine Model of Ischemic Stroke. Stroke 2018; 49:1248-1256. [PMID: 29650593 PMCID: PMC5916046 DOI: 10.1161/strokeaha.117.020353] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/28/2018] [Accepted: 03/12/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Recent work from our group suggests that human neural stem cell-derived extracellular vesicle (NSC EV) treatment improves both tissue and sensorimotor function in a preclinical thromboembolic mouse model of stroke. In this study, NSC EVs were evaluated in a pig ischemic stroke model, where clinically relevant end points were used to assess recovery in a more translational large animal model. METHODS Ischemic stroke was induced by permanent middle cerebral artery occlusion (MCAO), and either NSC EV or PBS treatment was administered intravenously at 2, 14, and 24 hours post-MCAO. NSC EV effects on tissue level recovery were evaluated via magnetic resonance imaging at 1 and 84 days post-MCAO. Effects on functional recovery were also assessed through longitudinal behavior and gait analysis testing. RESULTS NSC EV treatment was neuroprotective and led to significant improvements at the tissue and functional levels in stroked pigs. NSC EV treatment eliminated intracranial hemorrhage in ischemic lesions in NSC EV pigs (0 of 7) versus control pigs (7 of 8). NSC EV-treated pigs exhibited a significant decrease in cerebral lesion volume and decreased brain swelling relative to control pigs 1-day post-MCAO. NSC EVs significantly reduced edema in treated pigs relative to control pigs, as assessed by improved diffusivity through apparent diffusion coefficient maps. NSC EVs preserved white matter integrity with increased corpus callosum fractional anisotropy values 84 days post-MCAO. Behavior and mobility improvements paralleled structural changes as NSC EV-treated pigs exhibited improved outcomes, including increased exploratory behavior and faster restoration of spatiotemporal gait parameters. CONCLUSIONS This study demonstrated for the first time that in a large animal model novel NSC EVs significantly improved neural tissue preservation and functional levels post-MCAO, suggesting NSC EVs may be a paradigm changing stroke therapeutic.
Collapse
Affiliation(s)
- Robin L Webb
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S).,Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Erin E Kaiser
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice).,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences (E.E.K., F.D.W.)
| | - Brian J Jurgielewicz
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Samantha Spellicy
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| | - Shelley L Scoville
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S)
| | - Tyler A Thompson
- From the ArunA Biomedical, Athens, GA (R.L.W., S.L. Scoville, T.A.T., R.L.S)
| | | | - David C Hess
- University of Georgia, Rhodes Center for Animal and Dairy Science, Athens; and Department of Neurology, Augusta University, GA (D.C.H.)
| | - Franklin D West
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice).,Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences (E.E.K., F.D.W.)
| | - Steven L Stice
- Regenerative Bioscience Center (R.L.W., E.E.K., B.J.J., S.S., F.D.W., S.L. Stice)
| |
Collapse
|
288
|
Peak TC, Praharaj PP, Panigrahi GK, Doyle M, Su Y, Schlaepfer IR, Singh R, Vander Griend DJ, Alickson J, Hemal A, Atala A, Deep G. Exosomes secreted by placental stem cells selectively inhibit growth of aggressive prostate cancer cells. Biochem Biophys Res Commun 2018; 499:1004-1010. [PMID: 29627574 DOI: 10.1016/j.bbrc.2018.04.038] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 04/04/2018] [Indexed: 01/08/2023]
Abstract
The current paradigm in the development of new cancer therapies is the ability to target tumor cells while avoiding harm to noncancerous cells. Furthermore, there is a need to develop novel therapeutic options against drug-resistant cancer cells. Herein, we characterized the placental-derived stem cell (PLSC) exosomes (PLSCExo) and evaluated their anti-cancer efficacy in prostate cancer (PCa) cell lines. Nanoparticle tracking analyses revealed the size distribution (average size 131.4 ± 0.9 nm) and concentration of exosomes (5.23 × 1010±1.99 × 109 per ml) secreted by PLSC. PLSCExo treatment strongly inhibited the viability of enzalutamide-sensitive and -resistant PCa cell lines (C4-2B, CWR-R1, and LNCaP cells). Interestingly, PLSCExo treatment had no effect on the viability of a non-neoplastic human prostate cell line (PREC-1). Mass spectrometry (MS) analyses showed that PLSCExo are loaded with 241 proteins and mainly with saturated fatty acids. Further, Ingenuity Pathway Analysis analyses of proteins loaded in PLSCExo suggested the role of retinoic acid receptor/liver x receptor pathways in their biological effects. Together, these results suggest the novel selective anti-cancer effects of PLSCExo against aggressive PCa cells.
Collapse
Affiliation(s)
- Taylor C Peak
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Prakash P Praharaj
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Gati K Panigrahi
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Michael Doyle
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Yixin Su
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Isabel R Schlaepfer
- Division of Medical Oncology, Genitourinary Cancer Program, University of Colorado School of Medicine, Aurora, CO, United States
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Donald J Vander Griend
- Department of Surgery, Section of Urology, The University of Chicago, Chicago, IL, United States
| | - Julie Alickson
- Wake Forest Institute for Regenerative Medicine, United States
| | - Ashok Hemal
- Wake Forest Institute for Regenerative Medicine, United States; Department of Urology, Wake Forest School of Medicine, United States; Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, United States
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, United States; Department of Urology, Wake Forest School of Medicine, United States
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, United States; Department of Urology, Wake Forest School of Medicine, United States; Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, United States.
| |
Collapse
|
289
|
Rodrigues M, Fan J, Lyon C, Wan M, Hu Y. Role of Extracellular Vesicles in Viral and Bacterial Infections: Pathogenesis, Diagnostics, and Therapeutics. Am J Cancer Res 2018; 8:2709-2721. [PMID: 29774070 PMCID: PMC5957004 DOI: 10.7150/thno.20576] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 01/15/2018] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs), or exosomes, are nanovesicles of endocytic origin that carry host and pathogen-derived protein, nucleic acid, and lipid cargos. They are secreted by most cell types and play important roles in normal cell-to-cell communications but can also spread pathogen- and host-derived molecules during infections to alter immune responses and pathophysiological processes. New research is beginning to decipher how EVs influence viral and bacterial pathogenesis. In this review, we will describe how EVs influence viral and bacterial pathogenesis by spreading pathogen-derived factors and how they can promote and inhibit the immune response to these pathogens. We will also discuss the emerging potential of EVs as diagnostic and therapeutic tools.
Collapse
|
290
|
Li K, Rodosthenous RS, Kashanchi F, Gingeras T, Gould SJ, Kuo LS, Kurre P, Lee H, Leonard JN, Liu H, Lombo TB, Momma S, Nolan JP, Ochocinska MJ, Pegtel DM, Sadovsky Y, Sánchez-Madrid F, Valdes KM, Vickers KC, Weaver AM, Witwer KW, Zeng Y, Das S, Raffai RL, Howcroft TK. Advances, challenges, and opportunities in extracellular RNA biology: insights from the NIH exRNA Strategic Workshop. JCI Insight 2018; 3:98942. [PMID: 29618663 DOI: 10.1172/jci.insight.98942] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Extracellular RNA (exRNA) has emerged as an important transducer of intercellular communication. Advancing exRNA research promises to revolutionize biology and transform clinical practice. Recent efforts have led to cutting-edge research and expanded knowledge of this new paradigm in cell-to-cell crosstalk; however, gaps in our understanding of EV heterogeneity and exRNA diversity pose significant challenges for continued development of exRNA diagnostics and therapeutics. To unravel this complexity, the NIH convened expert teams to discuss the current state of the science, define the significant bottlenecks, and brainstorm potential solutions across the entire exRNA research field. The NIH Strategic Workshop on Extracellular RNA Transport helped identify mechanistic and clinical research opportunities for exRNA biology and provided recommendations on high priority areas of research that will advance the exRNA field.
Collapse
Affiliation(s)
- Kang Li
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, California, USA
| | | | - Fatah Kashanchi
- Laboratory of Molecular Virology, National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, USA
| | - Thomas Gingeras
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Stephen J Gould
- Department of Biological Chemistry, Johns Hopkins University, Baltimore, Maryland, USA
| | - Lillian S Kuo
- National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Peter Kurre
- Doernbecher Children's Hospital, Department of Pediatrics and Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Joshua N Leonard
- Department of Chemical and Biological Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA
| | - Huiping Liu
- Departments of Pharmacology and Medicine (Hematology and Oncology), Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tania B Lombo
- NIH, Office of the Director, Environmental Influences on Child Health Outcomes Program, Bethesda, Maryland, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), Frankfurt University Medical School, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Frankfurt, Heidelberg, Germany
| | - John P Nolan
- Scintillon Institute, San Diego, California, USA
| | | | - D Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Microbiology and Molecular Genetics, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria Princesa, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Kayla M Valdes
- National Center for Advancing Translational Science, Bethesda, Maryland, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alissa M Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kenneth W Witwer
- Department of Molecular and Comparative Pathobiology, Department of Neurology, The Johns Hopkins University, Baltimore, Maryland, USA
| | - Yong Zeng
- Department of Chemistry, University of Kansas Cancer Center, Lawrence, Kansas, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert L Raffai
- Division of Vascular and Endovascular Surgery, Department of Surgery, University of California, San Francisco, and Veterans Affairs Medical Center, San Francisco, California, USA
| | - T Kevin Howcroft
- Cancer Immunology, Hematology, and Etiology Branch, Division of Cancer Biology, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
291
|
Fuhrmann G, Chandrawati R, Parmar PA, Keane TJ, Maynard SA, Bertazzo S, Stevens MM. Engineering Extracellular Vesicles with the Tools of Enzyme Prodrug Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706616. [PMID: 29473230 PMCID: PMC5901706 DOI: 10.1002/adma.201706616] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/14/2017] [Indexed: 05/26/2023]
Abstract
Extracellular vesicles (EVs) have recently gained significant attention as important mediators of intercellular communication, potential drug carriers, and disease biomarkers. These natural cell-derived nanoparticles are postulated to be biocompatible, stable under physiological conditions, and to show reduced immunogenicity as compared to other synthetic nanoparticles. Although initial clinical trials are ongoing, the use of EVs for therapeutic applications may be limited due to undesired off-target activity and potential "dilution effects" upon systemic administration which may affect their ability to reach their target tissues. To fully exploit their therapeutic potential, EVs are embedded into implantable biomaterials designed to achieve local delivery of therapeutics taking advantage of enzyme prodrug therapy (EPT). In this first application of EVs for an EPT approach, EVs are used as smart carriers for stabilizing enzymes in a hydrogel for local controlled conversion of benign prodrugs to active antiinflammatory compounds. It is shown that the natural EVs' antiinflammatory potential is comparable or superior to synthetic carriers, in particular upon repeated long-term incubations and in different macrophage models of inflammation. Moreover, density-dependent color scanning electron microscopy imaging of EVs in a hydrogel is presented herein, an impactful tool for further understanding EVs in biological settings.
Collapse
Affiliation(s)
- Gregor Fuhrmann
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Biogenic Nanotherapeutics Group, Campus E8.1, 66123, Saarbrücken, Germany
| | - Rona Chandrawati
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Paresh A Parmar
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Timothy J Keane
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Stephanie A Maynard
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Sergio Bertazzo
- Department of Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, London, WC1E 6BT, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
292
|
Abstract
Large bone defects remain a tremendous clinical challenge. There is growing evidence in support of treatment strategies that direct defect repair through an endochondral route, involving a cartilage intermediate. While culture-expanded stem/progenitor cells are being evaluated for this purpose, these cells would compete with endogenous repair cells for limited oxygen and nutrients within ischaemic defects. Alternatively, it may be possible to employ extracellular vesicles (EVs) secreted by culture-expanded cells for overcoming key bottlenecks to endochondral repair, such as defect vascularization, chondrogenesis, and osseous remodelling. While mesenchymal stromal/stem cells are a promising source of therapeutic EVs, other donor cells should also be considered. The efficacy of an EV-based therapeutic will likely depend on the design of companion scaffolds for controlled delivery to specific target cells. Ultimately, the knowledge gained from studies of EVs could one day inform the long-term development of synthetic, engineered nanovesicles. In the meantime, EVs harnessed from in vitro cell culture have near-term promise for use in bone regenerative medicine. This narrative review presents a rationale for using EVs to improve the repair of large bone defects, highlights promising cell sources and likely therapeutic targets for directing repair through an endochondral pathway, and discusses current barriers to clinical translation. Cite this article: E. Ferreira, R. M. Porter. Harnessing extracellular vesicles to direct endochondral repair of large bone defects. Bone Joint Res 2018;7:263-273. DOI: 10.1302/2046-3758.74.BJR-2018-0006.
Collapse
Affiliation(s)
- E. Ferreira
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - R. M. Porter
- Departments of Internal Medicine and Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
293
|
Li Y, Cheng Q, Hu G, Deng T, Wang Q, Zhou J, Su X. Extracellular vesicles in mesenchymal stromal cells: A novel therapeutic strategy for stroke. Exp Ther Med 2018; 15:4067-4079. [PMID: 29725359 PMCID: PMC5920496 DOI: 10.3892/etm.2018.5993] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/18/2018] [Indexed: 12/11/2022] Open
Abstract
A stroke is a focal cerebral insult that frequently causes severe neurological deficit and mortality. Recent studies have demonstrated that multipotent mesenchymal stromal cells (MSCs) hold great promise for neurovascular remodeling and neurological function recovery following a stroke. Rather than a direct replacement of parenchymal brain cells, the therapeutic mechanism of MSCs is suggested to be the secretion of soluble factors. Specifically, emerging data described MSCs as being able to release extracellular vesicles (EVs), which contain a variety of cargo including proteins, lipids, DNA and various RNA species. The released EVs can target neurocytes and vascular cells and modify the cell's functions by delivering the cargo, which are considered to mediate the neural restoration effects of MSCs. Therefore, EVs may be developed as a novel cell-free therapy for neurological disorders. In the present review, the current advances regarding the components, functions and therapeutic potential of EVs are summarized and the use of MSC-derived EVs as a promising approach in the treatment of stroke are highlighted.
Collapse
Affiliation(s)
- Yingchen Li
- Post-doctoral Research Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Qilai Cheng
- College of Pharmacy, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Guoheng Hu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Tianhao Deng
- Department of Oncology, The Affiliated Hospital of Hunan Institute of Traditional Chinese Medicine, Changsha, Hunan 410006, P.R. China
| | - Qimei Wang
- Department of Oncology, The Affiliated Hospital of Hunan Institute of Traditional Chinese Medicine, Changsha, Hunan 410006, P.R. China
| | - Jianda Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xinping Su
- Department of Osteology, The Affiliated Hospital of Hunan Institute of Traditional Chinese Medicine, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
294
|
Mentkowski KI, Snitzer JD, Rusnak S, Lang JK. Therapeutic Potential of Engineered Extracellular Vesicles. AAPS JOURNAL 2018; 20:50. [PMID: 29546642 PMCID: PMC8299397 DOI: 10.1208/s12248-018-0211-z] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/27/2018] [Indexed: 12/16/2022]
Abstract
Extracellular vesicles (EVs) comprise a heterogeneous group of small membrane vesicles, including exosomes, which play a critical role in intracellular communication and regulation of numerous physiological processes in health and disease. Naturally released from virtually all cells, these vesicles contain an array of nucleic acids, lipids and proteins which they transfer to target cells within their local milieu and systemically. They have been proposed as a means of “cell-free, cell therapy” for cancer, immune disorders, and more recently cardiovascular disease. In addition, their unique properties of stability, biocompatibility, and low immunogenicity have prompted research into their potential as therapeutic delivery agents for drugs and small molecules. In this review, we aim to provide a comprehensive overview of the current understanding of extracellular vesicle biology as well as engineering strategies in play to improve their therapeutic potential.
Collapse
Affiliation(s)
- Kyle I Mentkowski
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Jonathan D Snitzer
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sarah Rusnak
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA
| | - Jennifer K Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical Sciences, Clinical and Translational Research Center, 895 Ellicott Street, Buffalo, NY, 14203, USA.
| |
Collapse
|
295
|
Shao H, Im H, Castro CM, Breakefield X, Weissleder R, Lee H. New Technologies for Analysis of Extracellular Vesicles. Chem Rev 2018; 118:1917-1950. [PMID: 29384376 PMCID: PMC6029891 DOI: 10.1021/acs.chemrev.7b00534] [Citation(s) in RCA: 1091] [Impact Index Per Article: 155.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are diverse, nanoscale membrane vesicles actively released by cells. Similar-sized vesicles can be further classified (e.g., exosomes, microvesicles) based on their biogenesis, size, and biophysical properties. Although initially thought to be cellular debris, and thus under-appreciated, EVs are now increasingly recognized as important vehicles of intercellular communication and circulating biomarkers for disease diagnoses and prognosis. Despite their clinical potential, the lack of sensitive preparatory and analytical technologies for EVs poses a barrier to clinical translation. New analytical platforms including molecular ones are thus actively being developed to address these challenges. Recent advances in the field are expected to have far-reaching impact in both basic and translational studies. This article aims to present a comprehensive and critical overview of emerging analytical technologies for EV detection and their clinical applications.
Collapse
Affiliation(s)
- Huilin Shao
- Departments of Biomedical Engineering and Surgery, National University of Singapore
- Biomedical Institute for Global Health Research and Technology, National University of Singapore
- Institute of Molecular and Cell Biology, Agency for Science Technology and Research
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
| | - Cesar M. Castro
- Center for Systems Biology, Massachusetts General Hospital
- Department of Medicine, Massachusetts General Hospital
| | - Xandra Breakefield
- Department of Radiology, Massachusetts General Hospital
- Department of Neurology, Massachusetts General Hospital
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
- Department of Systems Biology, Harvard Medical School
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital
- Department of Radiology, Massachusetts General Hospital
| |
Collapse
|
296
|
Cooks T, Pateras IS, Jenkins LM, Patel KM, Robles AI, Morris J, Forshew T, Appella E, Gorgoulis VG, Harris CC. Mutant p53 cancers reprogram macrophages to tumor supporting macrophages via exosomal miR-1246. Nat Commun 2018; 9:771. [PMID: 29472616 PMCID: PMC5823939 DOI: 10.1038/s41467-018-03224-w] [Citation(s) in RCA: 370] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/30/2018] [Indexed: 02/06/2023] Open
Abstract
TP53 mutants (mutp53) are involved in the pathogenesis of most human cancers. Specific mutp53 proteins gain oncogenic functions (GOFs) distinct from the tumor suppressor activity of the wild-type protein. Tumor-associated macrophages (TAMs), a hallmark of solid tumors, are typically correlated with poor prognosis. Here, we report a non-cell-autonomous mechanism, whereby human mutp53 cancer cells reprogram macrophages to a tumor supportive and anti-inflammatory state. The colon cancer cells harboring GOF mutp53 selectively shed miR-1246-enriched exosomes. Uptake of these exosomes by neighboring macrophages triggers their miR-1246-dependent reprogramming into a cancer-promoting state. Mutp53-reprogammed TAMs favor anti-inflammatory immunosuppression with increased activity of TGF-β. These findings, associated with poor survival in colon cancer patients, strongly support a microenvironmental GOF role for mutp53 in actively engaging the immune system to promote cancer progression and metastasis. p53 gain of function mutants (mutp53) are involved in the pathogenesis of most human cancers. Here, the authors show that mutp53 regulates the tumor microenvironment by inducing the release of specific exosomes containing miR-1246 that once received by macrophages turns them into tumor supportive macrophages.
Collapse
Affiliation(s)
- Tomer Cooks
- Laboratory of Human Carcinogenesis, NCI-CCR, National Institutes of Health, Bethesda, 20892-4258, MD, USA
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 75 Mikras Asias St, Athens, GR-11527, Greece
| | - Lisa M Jenkins
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, 20892-4258, MD, USA
| | - Keval M Patel
- Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Ana I Robles
- Laboratory of Human Carcinogenesis, NCI-CCR, National Institutes of Health, Bethesda, 20892-4258, MD, USA
| | - James Morris
- Cancer Research UK, Cambridge Research Institute, Robinsons Way, Cambridge, CB2 0RE, UK
| | - Tim Forshew
- UCL Cancer Institute, Huntley St, Camden Town, London, WC1E 6DD, UK
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, 20892-4258, MD, USA
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National Kapodistrian University of Athens, 75 Mikras Asias St, Athens, GR-11527, Greece.,Biomedical Research Foundation of the Academy of Athens, 4 Soranou Ephessiou St., GR-11527, Athens, Greece.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health, Science Centre, Wilmslow Road, Manchester, M20 4QL, UK
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, NCI-CCR, National Institutes of Health, Bethesda, 20892-4258, MD, USA.
| |
Collapse
|
297
|
Lässer C, Jang SC, Lötvall J. Subpopulations of extracellular vesicles and their therapeutic potential. Mol Aspects Med 2018; 60:1-14. [PMID: 29432782 DOI: 10.1016/j.mam.2018.02.002] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 01/17/2018] [Accepted: 02/06/2018] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs), such as exosomes and microvesicles, have over the last 10-15 years been recognized to convey key messages in the molecular communication between cells. Indeed, EVs have the capacity to shuttle proteins, lipids, and nucleotides such as RNA between cells, leading to an array of functional changes in the recipient cells. Importantly, the EV secretome changes significantly in diseased cells and under conditions of cellular stress. More recently, it has become evident that the EV secretome is exceptionally diverse, with many different types of EVs being released by a single cell type, and these EVs can be described in terms of differences in density, molecular cargos, and morphology. This review will discuss the diversity of EVs, will introduce some suggestions for how to categorize them, and will propose how EVs and their subpopulations might be used for very different therapeutic purposes.
Collapse
Affiliation(s)
- Cecilia Lässer
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Su Chul Jang
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden; Codiak BioSciences, Cambridge, MA 02139, USA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Sweden.
| |
Collapse
|
298
|
Martínez-Ballesta MDC, García-Gomez P, Yepes-Molina L, Guarnizo AL, Teruel JA, Carvajal M. Plasma membrane aquaporins mediates vesicle stability in broccoli. PLoS One 2018; 13:e0192422. [PMID: 29420651 PMCID: PMC5805300 DOI: 10.1371/journal.pone.0192422] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/23/2018] [Indexed: 12/25/2022] Open
Abstract
The use of in vitro membrane vesicles is attractive because of possible applications in therapies. Here we aimed to compare the stability and functionality of plasma membrane vesicles extracted from control and salt-treated broccoli. The impact of the amount of aquaporins was related to plasma membrane osmotic water permeability and the stability of protein secondary structure. Here, we describe for first time an increase in plant aquaporins acetylation under high salinity. Higher osmotic water permeability in NaCl vesicles has been related to higher acetylation, upregulation of aquaporins, and a more stable environment to thermal denaturation. Based on our findings, we propose that aquaporins play an important role in vesicle stability.
Collapse
Affiliation(s)
- Maria del Carmen Martínez-Ballesta
- Aquaporin Group, Plant Nutrition Department, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo, Espinardo, Murcia, Spain
| | - Pablo García-Gomez
- Aquaporin Group, Plant Nutrition Department, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo, Espinardo, Murcia, Spain
| | - Lucía Yepes-Molina
- Aquaporin Group, Plant Nutrition Department, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo, Espinardo, Murcia, Spain
| | - Angel L. Guarnizo
- Aquaporin Group, Plant Nutrition Department, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo, Espinardo, Murcia, Spain
| | - José A. Teruel
- Departamento de Bioquímica y Biología Molecular A, Facultad de Veterinaria, Universidad de Murcia, Espinardo, Murcia, Spain
| | - Micaela Carvajal
- Aquaporin Group, Plant Nutrition Department, Centro de Edafología y Biología Aplicada del Segura (CEBAS-CSIC), Campus de Espinardo, Espinardo, Murcia, Spain
- * E-mail:
| |
Collapse
|
299
|
Kooijmans SAA, Gitz-Francois JJJM, Schiffelers RM, Vader P. Recombinant phosphatidylserine-binding nanobodies for targeting of extracellular vesicles to tumor cells: a plug-and-play approach. NANOSCALE 2018; 10:2413-2426. [PMID: 29334397 PMCID: PMC5795695 DOI: 10.1039/c7nr06966a] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/10/2017] [Indexed: 05/14/2023]
Abstract
Extracellular vesicles (EVs) are increasingly being recognized as candidate drug delivery systems due to their ability to functionally transfer biological cargo between cells. However, manipulation of targeting properties of EVs through engineering of the producer cells can be challenging and time-consuming. As a novel approach to confer tumor targeting properties to isolated EVs, we generated recombinant fusion proteins of nanobodies against the epidermal growth factor receptor (EGFR) fused to phosphatidylserine (PS)-binding domains of lactadherin (C1C2). C1C2-nanobody fusion proteins were expressed in HEK293 cells and isolated from culture medium with near-complete purity as determined by SDS-PAGE. Fusion proteins specifically bound PS and showed no affinity for other common EV membrane lipids. Furthermore, C1C2 fused to anti-EGFR nanobodies (EGa1-C1C2) bound EGFR with high affinity and competed with binding of its natural ligand EGF, as opposed to C1C2 fused to non-targeting control nanobodies (R2-C1C2). Both proteins readily self-associated onto membranes of EVs derived from erythrocytes and Neuro2A cells without affecting EV size and integrity. EV-bound R2-C1C2 did not influence EV-cell interactions, whereas EV-bound EGa1-C1C2 dose-dependently enhanced specific binding and uptake of EVs by EGFR-overexpressing tumor cells. In conclusion, we developed a novel strategy to efficiently and universally confer tumor targeting properties to PS-exposing EVs after their isolation, without affecting EV characteristics, circumventing the need to modify EV-secreting cells. This strategy may also be employed to decorate EVs with other moieties, including imaging probes or therapeutic proteins.
Collapse
Affiliation(s)
- Sander A. A. Kooijmans
- Department of Clinical Chemistry and Haematology , University Medical Center Utrecht , Utrecht , The Netherlands . ; Tel: (+31) (0)887555546
| | - Jerney J. J. M. Gitz-Francois
- Department of Clinical Chemistry and Haematology , University Medical Center Utrecht , Utrecht , The Netherlands . ; Tel: (+31) (0)887555546
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology , University Medical Center Utrecht , Utrecht , The Netherlands . ; Tel: (+31) (0)887555546
| | - Pieter Vader
- Department of Clinical Chemistry and Haematology , University Medical Center Utrecht , Utrecht , The Netherlands . ; Tel: (+31) (0)887555546
| |
Collapse
|
300
|
Chaudhuri S, Korten T, Korten S, Milani G, Lana T, Te Kronnie G, Diez S. Label-Free Detection of Microvesicles and Proteins by the Bundling of Gliding Microtubules. NANO LETTERS 2018; 18:117-123. [PMID: 29202578 DOI: 10.1021/acs.nanolett.7b03619] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Development of miniaturized devices for the rapid and sensitive detection of analyte is crucial for various applications across healthcare, pharmaceutical, environmental, and other industries. Here, we report on the detection of unlabeled analyte by using fluorescently labeled, antibody-conjugated microtubules in a kinesin-1 gliding motility assay. The detection principle is based on the formation of fluorescent supramolecular assemblies of microtubule bundles and spools in the presence of multivalent analytes. We demonstrate the rapid, label-free detection of CD45+ microvesicles derived from leukemia cells. Moreover, we employ our platform for the label-free detection of multivalent proteins at subnanomolar concentrations, as well as for profiling the cross-reactivity between commercially available secondary antibodies. As the detection principle is based on the molecular recognition between antigen and antibody, our method can find general application where it identifies any analyte, including clinically relevant microvesicles and proteins.
Collapse
Affiliation(s)
- Samata Chaudhuri
- B CUBE - Center for Molecular Bioengineering, TU Dresden , 01069 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics , 01307 Dresden, Germany
| | - Till Korten
- B CUBE - Center for Molecular Bioengineering, TU Dresden , 01069 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics , 01307 Dresden, Germany
| | - Slobodanka Korten
- B CUBE - Center for Molecular Bioengineering, TU Dresden , 01069 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics , 01307 Dresden, Germany
| | - Gloria Milani
- Department of Women's and Children's Health, University of Padova , 35128 Padova, Italy
| | - Tobia Lana
- Department of Women's and Children's Health, University of Padova , 35128 Padova, Italy
| | - Geertruy Te Kronnie
- Department of Women's and Children's Health, University of Padova , 35128 Padova, Italy
| | - Stefan Diez
- B CUBE - Center for Molecular Bioengineering, TU Dresden , 01069 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics , 01307 Dresden, Germany
| |
Collapse
|