1
|
Nelson HM, Konar GJ, Patton JG. Isolation and Characterization of Extracellular Vesicles to Activate Retina Regeneration. Methods Mol Biol 2025; 2848:135-150. [PMID: 39240521 DOI: 10.1007/978-1-0716-4087-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Mammals do not possess the ability to spontaneously repair or regenerate damaged retinal tissue. In contrast to teleost fish which are capable of retina regeneration through the action of Müller glia, mammals undergo a process of reactive gliosis and scarring that inhibits replacement of lost neurons. Thus, it is important to discover novel methods for stimulating mammalian Müller glia to dedifferentiate and produce progenitor cells that can replace lost retinal neurons. Inducing an endogenous regenerative pathway mediated by Müller glia would provide an attractive alternative to stem cell injections or gene therapy approaches. Extracellular vesicles (EVs) are now recognized to serve as a novel form of cell-cell communication through the transfer of cargo from donor to recipient cells or by the activation of signaling cascades in recipient cells. EVs have been shown to promote proliferation and regeneration raising the possibility that delivery of EVs could be a viable treatment for visual disorders. Here, we provide protocols to isolate EVs for use in retina regeneration experiments.
Collapse
Affiliation(s)
- Hannah M Nelson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Gregory J Konar
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - James G Patton
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
2
|
Zhang Y, Liu Z, Chopp M, Millman M, Li Y, Cepparulo P, Kemper A, Li C, Zhang L, Zhang ZG. Small extracellular vesicles derived from cerebral endothelial cells with elevated microRNA 27a promote ischemic stroke recovery. Neural Regen Res 2025; 20:224-233. [PMID: 38767487 PMCID: PMC11246145 DOI: 10.4103/nrr.nrr-d-22-01292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/22/2024] [Indexed: 05/22/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202501000-00030/figure1/v/2024-05-14T021156Z/r/image-tiff Axonal remodeling is a critical aspect of ischemic brain repair processes and contributes to spontaneous functional recovery. Our previous in vitro study demonstrated that exosomes/small extracellular vesicles (sEVs) isolated from cerebral endothelial cells (CEC-sEVs) of ischemic brain promote axonal growth of embryonic cortical neurons and that microRNA 27a (miR-27a) is an elevated miRNA in ischemic CEC-sEVs. In the present study, we investigated whether normal CEC-sEVs engineered to enrich their levels of miR-27a (27a-sEVs) further enhance axonal growth and improve neurological outcomes after ischemic stroke when compared with treatment with non-engineered CEC-sEVs. 27a-sEVs were isolated from the conditioned medium of healthy mouse CECs transfected with a lentiviral miR-27a expression vector. Small EVs isolated from CECs transfected with a scramble vector (Scra-sEVs) were used as a control. Adult male mice were subjected to permanent middle cerebral artery occlusion and then were randomly treated with 27a-sEVs or Scra-sEVs. An array of behavior assays was used to measure neurological function. Compared with treatment of ischemic stroke with Scra-sEVs, treatment with 27a-sEVs significantly augmented axons and spines in the peri-infarct zone and in the corticospinal tract of the spinal grey matter of the denervated side, and significantly improved neurological outcomes. In vitro studies demonstrated that CEC-sEVs carrying reduced miR-27a abolished 27a-sEV-augmented axonal growth. Ultrastructural analysis revealed that 27a-sEVs systemically administered preferentially localized to the pre-synaptic active zone, while quantitative reverse transcription-polymerase chain reaction and Western Blot analysis showed elevated miR-27a, and reduced axonal inhibitory proteins Semaphorin 6A and Ras Homolog Family Member A in the peri-infarct zone. Blockage of the Clathrin-dependent endocytosis pathway substantially reduced neuronal internalization of 27a-sEVs. Our data provide evidence that 27a-sEVs have a therapeutic effect on stroke recovery by promoting axonal remodeling and improving neurological outcomes. Our findings also suggest that suppression of axonal inhibitory proteins such as Semaphorin 6A may contribute to the beneficial effect of 27a-sEVs on axonal remodeling.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Zhongwu Liu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Michael Millman
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Yanfeng Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | | - Amy Kemper
- Department of Pathology, Henry Ford Hospital, Detroit, MI, USA
| | - Chao Li
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | | |
Collapse
|
3
|
Gu M, Xu X, Wang X, Wang Y, Zhao Y, Hu X, Zhu L, Deng Z, Han C. Target Ligand Separation and Identification of Isoforsythiaside as a Histone Lysine-Specific Demethylase 1 Covalent Inhibitor Against Breast Cancer Metastasis. J Med Chem 2024. [PMID: 39499621 DOI: 10.1021/acs.jmedchem.4c02277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Histone lysine-specific demethylase 1 (LSD1) is hyperactive in breast cancer, which is associated with the metastasis of the tumor. Current irreversible LSD1 inhibitors are all synthesized by covalently binding to the flavin adenine dinucleotide cofactor, which often have side effects due to the high affinity for a variety of targets. Here, we identified isoforsythiaside (IFA), a natural phenylpropanoid glycoside isolated from Forsythia suspensa, as a novel covalent inhibitor of LSD1. The target ligand fishing technique and LC-MS/MS analysis identified that IFA could covalently bind to the Ser817 residue of LSD1 by α,β-unsaturated ketone moiety to block the amine oxidase-like domain of LSD1. Moreover, RBMS3/Twist1/MMP2, the downstream signaling pathway of LSD1, was activated after IFA treatment to inhibit the metastasis of MDA-MB-231 cells in vitro and in vivo. This study provided novel molecular templates for development of LSD1 covalence-binding inhibitor and laid a foundation for developing agents against breast carcinoma metastasis for targeting LSD1.
Collapse
Affiliation(s)
- Mengzhen Gu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoqing Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoping Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yun Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoxian Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lu Zhu
- Institute of Leisure Agriculture, Jiangsu Academy of Agricultural Sciences, 50 Zhongling Street, Nanjing 210014, China
| | - Zhenzhong Deng
- Department of Oncology, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, China
| | - Chao Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
4
|
Xu F, Chen M, Lin Y, Zhou S, Li J, Yu Y, Xu J, Wu W, Chen Y, Zhang H, Wei Y, Wang W. Functional Three-Dimensional Zeolitic Imidazolate Framework with an Ordered Macroporous Structure for the Isolation of Extracellular Vesicles. Anal Chem 2024; 96:17640-17648. [PMID: 39440634 DOI: 10.1021/acs.analchem.4c03566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) and their cargoes are increasingly being recognized as noninvasive diagnostic markers, necessitating the isolation of EVs from complex biological samples. Herein, a distearoyl phospholipid ethanolamine-functionalized single-crystal ordered macroporous three-dimensional zeolitic imidazolate framework (SOM-ZIF-8-DSPE) was developed, which combines the surface charge interaction of ZIF-8 with the synergistic effect of DSPE insertion into the phospholipid membrane of EVs to improve the isolating selectivity of EV capture. The materials have porous structures larger than 300 nm in diameter, providing enough space and active sites to trap EVs. Benefiting from this feature, the entire isolation process takes only 10 min and is well compatible with the subsequent analysis of RNA in EVs. Consequently, 10 upregulated miRNA of plasma EVs in the primary colorectal cancer (pCRC) patients is found over the healthy donors, and 6 upregulated miRNA of plasma EVs in the metastatic colorectal cancer (mCRC) patients over pCRC patients. These findings suggest that the isolation of EV-based SOM-ZIF-8-DSPE is a promising strategy to identify biomarkers for disease diagnosis, such as miRNAs in plasma EVs for the early detection of CRC.
Collapse
Affiliation(s)
- Fang Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mengxi Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yujie Lin
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Shenyue Zhou
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiaxi Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yuanyuan Yu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Jiayu Xu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Wen Wu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yinshuang Chen
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Haiyang Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yinmao Wei
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an 710127, China
| | - Weipeng Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
5
|
Lin J, Liu S, Xue X, Lv J, Zhao L, Yu L, Wang H, Chen J. Injectable Genetic Engineering Hydrogel for Promoting Spatial Tolerance of Transplanted Kidney in Situ. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408631. [PMID: 39498870 DOI: 10.1002/advs.202408631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/23/2024] [Indexed: 11/07/2024]
Abstract
The establishment of a tolerant space to realize the co-stimulation of cytokines and contact-dependent molecules remain challenging in allotransplant. Here, an injectable genetically engineered hydrogel (iGE-Gel) is reported, which developed with a multivalent network of FOXP3 engineered extracellular vesicles (Foe-EVs) through the hydrophobic interaction between stearic acid modified hyaluronic acid (HASA) and the membrane phospholipids of extracellular vesicles (EVs). The iGE-Gel exhibited self-healing properties, injectability and biocompatibility. It is revealed that iGE-Gel displayed with abundant regulatory cytokines and coinhibitory contact molecules, promoting the formation of immune tolerance in situ. The multiplex immunohistofluorescence confirmed tolerant niches is dominated by FOXP3+ Tregs and PDL1+ cells in the allograft, which reduced the drainage of alloantigens to subcapsular sinus of lymph nodes, and suppressed the formation of germinal centers. Remarkably, the proportion of alloreactive T cells (IFN-γ/IL-2) and B cells (IgG1/IgG2a/IgG3) as well as the serum titers of donor specific antibody (DSA) is decreased by iGE-Gel. In murine allogeneic transplantation, the injection of iGE-Gel significantly alleviated immune cell infiltration and complement damage in the graft, preserved the structure and function of renal cells and prolonged recipient survival period from 30.8 to 79.3 days, highlighting the potential of iGE-Gel as a transformative treatment in allotransplant.
Collapse
Affiliation(s)
- Jinwen Lin
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Shuaihui Liu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Xing Xue
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
| | - Junhao Lv
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Liqin Yu
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Huiping Wang
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Key Laboratory of Kidney Disease Prevention and Control Technology, National Key Clinical Department of Kidney Diseases, Institute of Nephrology, Zhejiang University, Zhejiang Clinical Research Center of Kidney and Urinary System Disease, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
6
|
Jiao H, Yu L. Migrasomes: Biogenesis, physiological roles, and therapeutic potentials. J Cell Biol 2024; 223:e202403051. [PMID: 39400310 PMCID: PMC11473597 DOI: 10.1083/jcb.202403051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Migrasomes, vesicular structures discovered in migrating cells, arise from the junctions or tips of retraction fibers, and gradually grow to microscale vesicles. Migrasomes have garnered attention for their role in intercellular communication and potential therapeutic implications. This review presents an overview of recent advances in migrasome biology, covering the mechanisms of migrasome biogenesis, essential physiological roles, and their association with various diseases, alongside potential therapeutic applications. Furthermore, we share our perspectives on potential future directions in the study of migrasomes and highlight the challenges that remain in this developing area of research.
Collapse
Affiliation(s)
- Haifeng Jiao
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Centre for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Dias C, Ballout N, Morla G, Alileche K, Santiago C, Guerrera IC, Chaubet A, Ausseil J, Trudel S. Extracellular vesicles from microglial cells activated by abnormal heparan sulfate oligosaccharides from Sanfilippo patients impair neuronal dendritic arborization. Mol Med 2024; 30:197. [PMID: 39497064 PMCID: PMC11536927 DOI: 10.1186/s10020-024-00953-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/07/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND In mucopolysaccharidosis type III (MPS III, also known as Sanfilippo syndrome), a pediatric neurodegenerative disorder, accumulation of abnormal glycosaminoglycans (GAGs) induces severe neuroinflammation by triggering the microglial pro-inflammatory cytokines production via a TLR4-dependent pathway. But the extent of the microglia contribution to the MPS III neuropathology remains unclear. Extracellular vesicles (EVs) mediate intercellular communication and are known to participate in the pathogenesis of adult neurodegenerative diseases. However, characterization of the molecular profiles of EVs released by MPS III microglia and their effects on neuronal functions have not been described. METHODS Here, we isolated EVs secreted by the microglial cells after treatment with GAGs purified from urines of Sanfilippo patients (sfGAGs-EVs) or from age-matched healthy subjects (nGAGs-EVs) to explore the EVs' proteins and small RNA profiles using LC-MS/MS and RNA sequencing. We next performed a functional assay by immunofluorescence following nGAGs- or sfGAGs-EVs uptake by WT primary cortical neurons and analyzed their extensions metrics after staining of βIII-tubulin and MAP2 by confocal microscopy. RESULTS Functional enrichment analysis for both proteomics and RNA sequencing data from sfGAGs-EVs revealed a specific content involved in neuroinflammation and neurodevelopment pathways. Treatment of cortical neurons with sfGAGs-EVs induced a disease-associated phenotype demonstrated by a lower total neurite surface area, an impaired somatodendritic compartment, and a higher number of immature dendritic spines. CONCLUSIONS This study shows, for the first time, that GAGs from patients with Sanfilippo syndrome can induce microglial secretion of EVs that deliver a specific molecular message to recipient naive neurons, while promoting the neuroinflammation, and depriving neurons of neurodevelopmental factors. This work provides a framework for further studies of biomarkers to evaluate efficiency of emerging therapies.
Collapse
Affiliation(s)
- Chloé Dias
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Nissrine Ballout
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Guillaume Morla
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Katia Alileche
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Christophe Santiago
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Ida Chiara Guerrera
- Necker Proteomics Platform, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR3633, Université Paris Cité, 75015, Paris, France
| | - Adeline Chaubet
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
| | - Jerome Ausseil
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France
- Laboratoire de Biochimie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stephanie Trudel
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Inserm U1291, CNRS U5051, University of Toulouse, Toulouse, France.
- Laboratoire de Biochimie, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.
| |
Collapse
|
8
|
Sies H. SFRRI Inaugural Alberto Boveris Award Lecture Dynamics of Intracellular and Intercellular Redox Communication. Free Radic Biol Med 2024:S0891-5849(24)01024-4. [PMID: 39491734 DOI: 10.1016/j.freeradbiomed.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/01/2024] [Indexed: 11/05/2024]
Abstract
Cell and organ metabolism is organized through various signaling mechanisms, including redox, Ca2+, kinase and electrochemical pathways. Redox signaling operates at multiple levels, from interactions between individual molecules in their microenvironment to communication among subcellular organelles, single cells, organs, and the entire organism. Redox communication is a dynamic and ongoing spatiotemporal process. This article focuses on hydrogen peroxide (H2O2), a key second messenger that targets redox-active protein cysteine thiolates. H2O2 gradients across cell membranes are controlled by peroxiporins, specialized aquaporins. Redox-active endosomes, known as redoxosomes, form at the plasma membrane. Cell-to-cell redox communication involves direct contacts, such as per gap junctions that connect cells for transfer of molecules via connexons. Moreover, signaling occurs through the release of redox-active molecules and enzymes into the surrounding space, as well as through various extracellular vesicles (EVs) that transport these signals to nearby or distant target cells.
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Medical Faculty, Heinrich-Heine-University Düsseldorf, Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| |
Collapse
|
9
|
Rayamajhi S, Gibbs BK, Sipes J, Pathak HB, Bossmann SH, Godwin AK. Tracking Small Extracellular Vesicles Using a Minimally Invasive PicoGreen Labeling Strategy. ACS APPLIED BIO MATERIALS 2024. [PMID: 39482871 DOI: 10.1021/acsabm.4c01500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Extracellular vesicles (EVs) are cell-secreted lipid bilayer delimited particles that mediate cellular communication. These tiny sacs of cellular information play an important role in cell communication and alter the physiological process under both normal and pathological conditions. As such, tracking EVs can provide valuable information regarding the basic understanding of cell communication, the onset of early malignancy, and biomarker discovery. Most of the current EV-tracking strategies are invasive, altering the natural characteristics of EVs by modifying the lipid bilayer with lipophilic dyes or surface proteins with fluorescent reporters. The invasive labeling strategies could alter the natural processes of EVs and thereby have major limitations for functional studies. Here, we report an alternative minimally invasive EV labeling strategy using PicoGreen (PG), a small molecule that fluoresces at 520 nm when bound to dsDNA. We show that PG binds to dsDNA associated with small EVs (50-200 nm), forming a stable and highly fluorescent PG-DNA complex in EVs (PG-EVs). In both 2D cell culture and 3D organoid models, PG-EV showed efficient tracking properties, including a high signal-to-noise ratio, time- and concentration-dependent uptake, and the ability to traverse a 3D environment. We further validated PG-EV tracking using dual-labeled EVs following two orthogonal labeling strategies: (1) Bioconjugation via surface amine labeling and (2) donor cell engineering via endogenously expressing mCherry-tetraspanin (CD9/CD63/CD81) reporter proteins. Our study has shown the feasibility of using PG-EV as an effective EV tracking strategy that can be applied for studying the functional role of EVs across multiple model systems.
Collapse
Affiliation(s)
- Sagar Rayamajhi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Benjamin K Gibbs
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Jared Sipes
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Stefan H Bossmann
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| |
Collapse
|
10
|
Chen J, Wang Z, Liu S, Zhao R, Chen Q, Li X, Zhang S, Wang J. Lymphocyte-Derived Engineered Apoptotic Bodies with Inflammation Regulation and Cartilage Affinity for Osteoarthritis Therapy. ACS NANO 2024; 18:30084-30098. [PMID: 39403980 DOI: 10.1021/acsnano.4c11622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Apoptotic bodies as plentiful extracellular vesicles generated from apoptotic cells play a central role in signal transduction and homeostasis regulation and simultaneously switch death to regeneration to a certain extent. Herein, we designed engineered apoptotic bodies derived from T cells to have the capacity of inflammation regulation and cartilage affinity. The engineered apoptotic bodies as a natural anti-inflammation factor were encapsulated into lubricating hydrogel microspheres to achieve an injectable microsphere complex for the treatment of osteoarthritis (OA). In the above therapeutic system, the engineered apoptotic bodies acted as a biochemical cue to regulate the inflammatory microenvironment and promote chondrocyte cartilage homeostasis, whereas the lubricating hydrogel microspheres served as a biophysical stimulation to effectively reduce the friction of the cartilage surface, restore the cartilage stress, and control the slow delivery of the encapsulated engineered apoptotic bodies by friction degradation. Consequently, the current work creates an injectable and multifunctional therapeutic microsphere to advance cartilage remodeling and OA therapy.
Collapse
Affiliation(s)
- Jia Chen
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zihao Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shuaibing Liu
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ruiyue Zhao
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qi Chen
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, National Center for International Joint Research of Micro-Nano Molding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jianglin Wang
- Department of Biomedical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- NMPA Research Base of Regulatory Science for Medical Devices, Institute of Regulatory Science for Medical Devices, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
11
|
Bader J, Brigger F, Leroux JC. Extracellular vesicles versus lipid nanoparticles for the delivery of nucleic acids. Adv Drug Deliv Rev 2024:115461. [PMID: 39490384 DOI: 10.1016/j.addr.2024.115461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Extracellular vesicles (EVs) are increasingly investigated for delivering nucleic acid (NA) therapeutics, leveraging their natural role in transporting NA and protein-based cargo in cell-to-cell signaling. Their synthetic counterparts, lipid nanoparticles (LNPs), have been developed over the past decades as NA carriers, culminating in the approval of several marketed formulations such as patisiran/Onpattro® and the mRNA-1273/BNT162 COVID-19 vaccines. The success of LNPs has sparked efforts to develop innovative technologies to target extrahepatic organs, and to deliver novel therapeutic modalities, such as tools for in vivo gene editing. Fueled by the recent advancements in both fields, this review aims to provide a comprehensive overview of the basic characteristics of EV and LNP-based NA delivery systems, from EV biogenesis to structural properties of LNPs. It addresses the primary challenges encountered in utilizing these nanocarriers from a drug formulation and delivery perspective. Additionally, biodistribution profiles, in vitro and in vivo transfection outcomes, as well as their status in clinical trials are compared. Overall, this review provides insights into promising research avenues and potential dead ends for EV and LNP-based NA delivery systems.
Collapse
Affiliation(s)
- Johannes Bader
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Finn Brigger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
12
|
Gu X, He L, Zhang J, Xu H, Shen H, Huang R, Li Z. Recent Advances in Wash-Free Detection Methods of Extracellular Vesicles: A Mini Review. ACS Sens 2024. [PMID: 39446112 DOI: 10.1021/acssensors.4c00315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are emerging biomarkers in liquid biopsy that have gained increasing attention in disease diagnosis and prognosis monitoring. Most reported detection methods require the isolation of EVs from complex body liquids, often involving multiple washing steps to remove excess reagents and eliminate background interference. Nonetheless, these methods not only cause the loss of EVs but also result in poor repeatability and prolonged detection duration. The focus on wash-free detection methods is increasing due to the specific ability to avoid the removal of surplus reagents and, in some cases, even the isolation and purification of EVs. Viewing from different methodological perspectives, this review summarizes the recent advances in wash-free detection of EVs, containing aggregation induction, proximity sensing, allosteric probes, phase separation, Roman spectroscopy, field-effect transistor and microcantilever. The pros and cons of each detection strategy are impartially evaluated and this review concludes the prospects for future developments in this field.
Collapse
Affiliation(s)
- Xinrui Gu
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Lei He
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Jinsong Zhang
- Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Hongpan Xu
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Han Shen
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, South Puzhu Road 30, Nanjing, Jiangsu Province 211816, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| |
Collapse
|
13
|
Gao K, Xi W, Ni J, Jiang J, Lei Y, Li L, Chu J, Li R, An Y, Ouyang Y, Su R, Zhang R, Wu G. Genetically modified extracellular vesicles loaded with activated gasdermin D potentially inhibit prostate-specific membrane antigen-positive prostate carcinoma growth and enhance immunotherapy. Biomaterials 2024; 315:122894. [PMID: 39461061 DOI: 10.1016/j.biomaterials.2024.122894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
Prostate cancer (PCa) is associated with poor immunogenicity and lymphocytic infiltration, and immunotherapy effective against PCa remains unavailable. Pyroptosis, a novel immunotherapeutic modality for cancer, promotes systemic immune responses leading to immunogenic cell death in solid tumors. This paper describes the preparation and analysis of PSMAscFv-EVN-GSDMD; this genetically engineered recombinant extracellular vesicle (EV) expresses a single-chain variable antibody fragment (scFv) with high affinity for prostate-specific membrane antigen (PSMA) on their surfaces and is loaded with the N-terminal domain of gasdermin D (GSDMD). Both in vitro and in vivo, PSMAscFv-EVN-GSDMD effectively targeted PSMA-positive PCa cells and induced pyroptosis through the carrier properties of EVs and the specificity of PSMAscFv. In the 22RV1 and PSMA-transfected RM-1-inoculated PCa mouse models, PSMAscFv-EVN-GSDMD efficiently inhibited tumor growth and promoted tumor immune responses. In conclusion, PSMAscFv-EVN-GSDMD can convert the immunosuppressive "cold" tumor microenvironment of PCa into an immunogenic "hot" tumor microenvironment.
Collapse
Affiliation(s)
- Ke Gao
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China; State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Wenjin Xi
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Jianxin Ni
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Jun Jiang
- Department of Health Service, Base of Health Service, Air Force Medical University, Xi'an, China
| | - Yonghua Lei
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Lin Li
- Department of Immunology, School of Basic Medicine, Yan'an University. Yan'an, 716099, China
| | - Jie Chu
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China
| | - Ruixiao Li
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Yongpan An
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Yanan Ouyang
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Ruiping Su
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Rui Zhang
- State Key Laboratory of Cancer Biology, Department of Immunology, Air Force Medical University, Xi'an, Shaanxi, 710032, China; State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China.
| | - Guojun Wu
- Department of Urology, Xi'an People's Hospital (Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China.
| |
Collapse
|
14
|
Mkrtchian S, Eldh M, Ebberyd A, Gabrielsson S, Végvári Á, Ricksten SE, Danielson M, Oras J, Wiklund A, Eriksson LI, Gómez-Galán M. Changes in circulating extracellular vesicle cargo are associated with cognitive decline after major surgery: an observational case-control study. Br J Anaesth 2024:S0007-0912(24)00553-1. [PMID: 39426921 DOI: 10.1016/j.bja.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/27/2024] [Accepted: 07/21/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Postoperative neurocognitive decline is a frequent complication triggered by unclear signalling mechanisms. This observational case-control study investigated the effects of hip or knee replacement surgery on the composition of circulating extracellular vesicles (EVs), potential periphery-to-brain messengers, and their association with neurocognitive outcomes. METHODS We mapped the microRNAome and proteome of plasma-derived EVs from 12 patients (six with good and six with poor neurocognitive outcomes at 3 months after surgery) at preoperative and postoperative timepoints (4, 8, 24, and 48 h). Complement C3-EV association was confirmed by flow cytometry in plasma- and cerebrospinal fluid (CSF)-derived EVs, with total plasma and CSF C3 and C3a concentrations determined using enzyme-linked immunosorbent assay. RESULTS Differential expression analysis found eight dysregulated EV microRNAs (miRNAs) exclusively in the poor neurocognitive outcomes group. Pathway analysis suggested potential downregulation of proliferative pathways and activation of extracellular matrix and inflammatory response pathways in EV target tissues. Proteome analysis revealed a time-dependent increase in immune-related EV proteins, including complement system proteins, notably EV surface-associated C3. Such upward kinetics was detected earlier in the poor neurocognitive outcomes group. Interestingly, CSF-derived EVs from the same group showed a drastic drop of C3 at 48 h with unchanged concentrations in the good neurocognitive outcomes group. Functionally, the complement system was activated in both patient groups in plasma, but only in the poor neurocognitive outcomes group in CSF. CONCLUSIONS Our findings highlight the impact of surgery on plasma- and CSF-derived EVs, particularly in patients with poor neurocognitive outcomes, indicating a potential role for EVs. The small sample size necessitates verification with a larger patient cohort.
Collapse
Affiliation(s)
- Souren Mkrtchian
- Department of Physiology and Pharmacology, Section for Anaesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eldh
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Anette Ebberyd
- Department of Physiology and Pharmacology, Section for Anaesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Gabrielsson
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medicinal Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Sven-Erik Ricksten
- Department of Anesthesia and Intensive Care, Sаhlgrenska University Hospital, Gothenburg, Sweden; Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Mattias Danielson
- Department of Anesthesia and Intensive Care, Sаhlgrenska University Hospital, Gothenburg, Sweden; Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jonatan Oras
- Department of Anesthesia and Intensive Care, Sаhlgrenska University Hospital, Gothenburg, Sweden; Department of Anesthesiology and Intensive Care Medicine, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Andreas Wiklund
- Department of Physiology and Pharmacology, Section for Anaesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden; Capio Artro Clinic, Stockholm, Sweden
| | - Lars I Eriksson
- Department of Physiology and Pharmacology, Section for Anaesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden; Function Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Marta Gómez-Galán
- Department of Physiology and Pharmacology, Section for Anaesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
15
|
Wang Y, Wang C, Yang F, Chen Y, Shi Y, Xu R, Zhang Z, Yan Y. USP9X-enriched MSC-sEV inhibits LSEC angiogenesis in MASH mice by downregulating the IκBα/NF-κB/Ang-2 pathway. Pharmacol Res 2024; 209:107471. [PMID: 39427871 DOI: 10.1016/j.phrs.2024.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
Pathological angiogenesis of liver sinusoidal endothelial cells (LSEC) plays a crucial role in the progression of metabolic dysfunction-associated steatohepatitis (MASH)-induced liver fibrosis. Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEV) have shown promising therapeutic potential against MASH. This study aimed to investigate the impact of MSC-sEV on LSEC angiogenesis and elucidate the underlying molecular mechanisms. The effects of MSC-sEV on LSEC angiogenesis were evaluated in Tumor Necrosis Factor- alpha (TNF-α)-treated LSECs in vitro and in Methionine and Choline Deficient Diet (MCD)-induced MASH mice in vivo. Herein, we found that MSC-sEV effectively suppressed LSEC angiogenesis by targeting the angiogenesis marker Angiogenin 2 (Ang-2) in both TNF-α-treated LSECs and MASH mice. Gene manipulation experiments revealed that the primary mechanism by which MSC-sEV inhibited LSEC angiogenesis was through the modulation of nuclear factor kappa B inhibitor alpha (IκBα) / nuclear factor kappa B (NF-κB) / Ang-2 pathway. Additionally, mass spectrometry and co-immunoprecipitation (Co-IP) data suggested that MSC-sEV delivered the ubiquitin specific peptidase 9 X-linked (USP9X) protein to LSECs, leading to enhanced IκBα deubiquitination and NF-κB in activation, ultimately resulting in the inhibition of Ang-2-mediated LSEC angiogenesis. Knockdown of USP9X attenuated the regulatory effects of MSC-sEV on Ang-2 expression, LSEC angiogenesis, and the progression of MASH. In conclusion, our findings indicate that USP9X delivered via MSC-sEV can suppress LSEC angiogenesis and alleviate MASH-induced liver fibrosis through the IκBα/NF-κB/Ang-2 signaling pathway.
Collapse
Affiliation(s)
- Yanjin Wang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Chen Wang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fuji Yang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yifei Chen
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yujie Shi
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ruizi Xu
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhuan Zhang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China.
| |
Collapse
|
16
|
Roldán Gallardo FF, Martínez Piñerez DE, Reinarz Torrado KF, Berg GA, Herzfeld JD, Da Ros VG, López Seoane M, Maldonado CA, Quintar AA. Extracellular Vesicles Contribute to Oxidized LDL-Induced Stromal Cell Proliferation in Benign Prostatic Hyperplasia. BIOLOGY 2024; 13:827. [PMID: 39452137 PMCID: PMC11504470 DOI: 10.3390/biology13100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Clinical and experimental evidence has linked Benign Prostatic Hyperplasia (BPH) with dyslipidemic and hypercholesterolemic conditions, though the underlying cellular mechanisms remain unclear. This study investigates the impact of dyslipidemia, specifically oxidized LDL (OxLDL), on prostatic stromal cell proliferation and the release of extracellular vesicles (EVs). METHODS Mice were fed a high-fat diet, and human prostatic stromal cells (HPSCs) were treated with OxLDL. Proliferation assays and EV characterization were performed to assess the role of EVs in BPH progression. RESULTS Pro-atherogenic conditions significantly increased cell proliferation in both murine prostatic cells and HPSCs. Treatment with metformin effectively inhibited OxLDL-induced proliferation. Additionally, OxLDL stimulated the production and release of pro-proliferative EVs by HPSCs, which further promoted cellular proliferation. CONCLUSIONS The findings suggest that dyslipidemia drives prostatic stromal cell proliferation and EV secretion, contributing to BPH progression. Metformin demonstrates potential as a therapeutic agent to mitigate these effects, offering insight into novel strategies for BPH management. This study highlights the complex interaction between dyslipidemia, cell proliferation, and extracellular communication in the context of BPH pathogenesis.
Collapse
Affiliation(s)
- Franco F. Roldán Gallardo
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Daniel E. Martínez Piñerez
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
| | - Kevin F. Reinarz Torrado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
| | - Gabriela A. Berg
- Departamento de Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires 1000, Argentina
| | - Jael D. Herzfeld
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1000, Argentina
| | - Vanina G. Da Ros
- Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires 1000, Argentina
| | | | - Cristina A. Maldonado
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| | - Amado A. Quintar
- Centro de Microscopía Electrónica, Facultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba 5000, Argentina; (F.F.R.G.)
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba 5000, Argentina
| |
Collapse
|
17
|
Maqueda JJ, De Feo A, Scotlandi K. Evaluating Circulating Biomarkers for Diagnosis, Prognosis, and Tumor Monitoring in Pediatric Sarcomas: Recent Advances and Future Directions. Biomolecules 2024; 14:1306. [PMID: 39456239 PMCID: PMC11506719 DOI: 10.3390/biom14101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Pediatric sarcomas present a significant challenge in oncology. There is an urgent need for improved therapeutic strategies for high-risk patients and better management of long-term side effects for those who survive the disease. Liquid biopsy is emerging as a promising tool to optimize treatment in these patients by offering non-invasive, repeatable assessments of disease status. Circulating biomarkers can provide valuable insights into tumor genetics and treatment response, potentially facilitating early diagnosis and dynamic disease monitoring. This review examines the potential of liquid biopsies, focusing on circulating biomarkers in the most common pediatric sarcomas, i.e., osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma. We also highlight the current research efforts and the necessary advancements required before these technologies can be widely adopted in clinical practice.
Collapse
Affiliation(s)
- Joaquín J. Maqueda
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (A.D.F.); (K.S.)
| | | | | |
Collapse
|
18
|
Gurrieri E, Carradori G, Roccuzzo M, Pancher M, Peroni D, Belli R, Trevisan C, Notarangelo M, Huang WQ, Carreira ASA, Quattrone A, Jenster G, Hagen TLMT, D'Agostino VG. CD81-guided heterologous EVs present heterogeneous interactions with breast cancer cells. J Biomed Sci 2024; 31:92. [PMID: 39402557 PMCID: PMC11475557 DOI: 10.1186/s12929-024-01084-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are cell-secreted particles conceived as natural vehicles for intercellular communication. The capacity to entrap heterogeneous molecular cargoes and target specific cell populations through EV functionalization promises advancements in biomedical applications. However, the efficiency of the obtained EVs, the contribution of cell-exposed receptors to EV interactions, and the predictability of functional cargo release with potential sharing of high molecular weight recombinant mRNAs are crucial for advancing heterologous EVs in targeted therapy applications. METHODS In this work, we selected the popular EV marker CD81 as a transmembrane guide for fusion proteins with a C-terminal GFP reporter encompassing or not Trastuzumab light chains targeting the HER2 receptor. We performed high-content imaging analyses to track EV-cell interactions, including isogenic breast cancer cells with manipulated HER2 expression. We validated the functional cargo delivery of recombinant EVs carrying doxorubicin upon EV-donor cell treatment. Then, we performed an in vivo study using JIMT-1 cells commonly used as HER2-refractory, trastuzumab-resistant model to detect a more than 2000 nt length recombinant mRNA in engrafted tumors. RESULTS Fusion proteins participated in vesicular trafficking dynamics and accumulated on secreted EVs according to their expression levels in HEK293T cells. Despite the presence of GFP, secreted EV populations retained a HER2 receptor-binding capacity and were used to track EV-cell interactions. In time-frames where the global EV distribution did not change between HER2-positive (SK-BR-3) or -negative (MDA-MB-231) breast cancer cell lines, the HER2 exposure in isogenic cells remarkably affected the tropism of heterologous EVs, demonstrating the specificity of antiHER2 EVs representing about 20% of secreted bulk vesicles. The specific interaction strongly correlated with improved cell-killing activity of doxorubicin-EVs in MDA-MB-231 ectopically expressing HER2 and reduced toxicity in SK-BR-3 with a knocked-out HER2 receptor, overcoming the effects of the free drug. Interestingly, the fusion protein-corresponding transcripts present as full-length mRNAs in recombinant EVs could reach orthotopic breast tumors in JIMT-1-xenografted mice, improving our sensitivity in detecting penetrant cargoes in tissue biopsies. CONCLUSIONS This study highlights the quantitative aspects underlying the creation of a platform for secreted heterologous EVs and shows the limits of single receptor-ligand interactions behind EV-cell engagement mechanisms, which now become the pivotal step to predict functional tropism and design new generations of EV-based nanovehicles.
Collapse
Affiliation(s)
- Elena Gurrieri
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Giulia Carradori
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Michela Roccuzzo
- Advanced Imaging Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Michael Pancher
- High Throughput Screening and High Content Analysis Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Daniele Peroni
- Mass Spectrometry and Proteomics Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Romina Belli
- Mass Spectrometry and Proteomics Core Facility, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Caterina Trevisan
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Michela Notarangelo
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Wen-Qiu Huang
- Precision Medicine in Oncology (PrMiO), Department of Pathology, and Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC Cancer Institute, 3015 GD, Rotterdam, The Netherlands
| | - Agata S A Carreira
- Laboratory of Genomic Screening, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy
| | - Guido Jenster
- Department of Urology, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), Department of Pathology, and Nanomedicine Innovation Center Erasmus (NICE), Erasmus MC Cancer Institute, 3015 GD, Rotterdam, The Netherlands
| | - Vito Giuseppe D'Agostino
- Laboratory of Biotechnology and Nanomedicine, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123, Trento, Italy.
| |
Collapse
|
19
|
Liu J, Verweij FJ, van Niel G, Galli T, Danglot L, Bun P. ExoJ - a Fiji/ImageJ2 plugin for automated spatiotemporal detection and analysis of exocytosis. J Cell Sci 2024; 137:jcs261938. [PMID: 39219469 DOI: 10.1242/jcs.261938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Exocytosis is a dynamic physiological process that enables the release of biomolecules to the surrounding environment via the fusion of membrane compartments to the plasma membrane. Understanding its mechanisms is crucial, as defects can compromise essential biological functions. The development of pH-sensitive optical reporters alongside fluorescence microscopy enables the assessment of individual vesicle exocytosis events at the cellular level. Manual annotation represents, however, a time-consuming task that is prone to selection biases and human operational errors. Here, we introduce ExoJ, an automated plugin based on Fiji/ImageJ2 software. ExoJ identifies user-defined genuine populations of exocytosis events, recording quantitative features including intensity, apparent size and duration. We designed ExoJ to be fully user-configurable, making it suitable for studying distinct forms of vesicle exocytosis regardless of the imaging quality. Our plugin demonstrates its capabilities by showcasing distinct exocytic dynamics among tetraspanins and vesicular SNARE protein reporters. Assessment of performance on synthetic data shows that ExoJ is a robust tool that is capable of correctly identifying exocytosis events independently of signal-to-noise ratio conditions. We propose ExoJ as a standard solution for future comparative and quantitative studies of exocytosis.
Collapse
Affiliation(s)
- Junjun Liu
- Jinan Central Hospital affiliated to Shandong First Medical University, Jinan 250013, China
| | | | - Guillaume van Niel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Endosomal dynamics in neuropathies, 75014 Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Saint Anne, F-75014 Paris, France
| | - Thierry Galli
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Saint Anne, F-75014 Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Membrane traffic in healthy and diseased brain, 75014 Paris, France
| | - Lydia Danglot
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Saint Anne, F-75014 Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Membrane traffic in healthy and diseased brain, 75014 Paris, France
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging Core Facility, 75014 Paris, France
| | - Philippe Bun
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, NeurImag Imaging Core Facility, 75014 Paris, France
| |
Collapse
|
20
|
Pierce C, Suryoraharjo K, Robertson IH, Su X, Hatchett DB, Shin A, Adams KN, Berthier E, Thongpang S, Ogata A, Theberge AB, Sohn LL. CandyCollect: An Open-Microfluidic Device for the Direct Capture and Enumeration of Salivary-Extracellular Vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617508. [PMID: 39463934 PMCID: PMC11507796 DOI: 10.1101/2024.10.09.617508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Extracellular Vesicles (EVs) are membrane-derived vesicles shed by cells into the extracellular space that play key roles in intercellular communication and other biological processes. As membrane-bound cargos of nucleic acids and other proteins that are abundantly found in virtually every biofluid including blood, urine, and saliva, EVs are widely regarded as promising biomarkers for disease detection. While it is an increasingly promising biofluid from which to isolate EVs, saliva poses challenges due its complexity and heterogeneity-cells, debris, and other proteins can inhibit the isolation of EVs by traditional platforms. Here, we employ the CandyCollect, a lollipop-inspired sampling device with open microfluidic channels, as a non-invasive and patient-friendly alternative for the capture of salivary EVs. The CandyCollect simplifies sample preparation by effectively pre-concentrating EVs on the device surface before EVs are eluted off of the CandyCollect, labeled with cholesterol-tagged oligonucleotides, and subsequently detected by qPCR with primers specific for the tagged oligos to enumerate the relative number of EVs. We demonstrate that downstream EV cargo analysis can be performed using Simoa. Overall, the CandyCollect ushers a new method to capture, enumerate, and analyze salivary EVs.
Collapse
|
21
|
Gurriaran-Rodriguez U, De Repentigny Y, Kothary R, Rudnicki MA. Isolation of small extracellular vesicles from regenerating muscle tissue using tangential flow filtration and size exclusion chromatography. Skelet Muscle 2024; 14:22. [PMID: 39394606 PMCID: PMC11468478 DOI: 10.1186/s13395-024-00355-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
We have recently made the strikingly discovery that upon a muscle injury, Wnt7a is upregulated and secreted from new regenerating myofibers on the surface of exosomes to elicit its myogenerative response distally. Despite recent advances in extracellular vesicle (EVs) isolation from diverse tissues, there is still a lack of specific methodology to purify EVs from muscle tissue. To eliminate contamination with non-EV secreted proteins and cytoplasmic fragments, which are typically found when using classical methodology, such as ultracentrifugation, we adapted a protocol combining Tangential Flow Filtration (TFF) and Size Exclusion Chromatography (SEC). We found that this approach allows simultaneous purification of Wnt7a, bound to EVs (retentate fraction) and free non-EV Wnt7a (permeate fraction). Here we described this optimized protocol designed to specifically isolate EVs from hind limb muscle explants, without cross-contamination with other sources of non-EV bounded proteins. The first step of the protocol is to remove large EVs with sequential centrifugation. Extracellular vesicles are then concentrated and washed in exchange buffer by TFF. Lastly, SEC is performed to remove any soluble protein traces remaining after TFF. Overall, this procedure can be used to isolate EVs from conditioned media or biofluid that contains EVs derived from any cell type or tissue, improving reproducibility, efficiency, and purity of EVs preparations. Our purification protocol results in high purity EVs that maintain structural integrity and thus fully compatible with in vitro and in vivo bioactivity and analytic assays.
Collapse
Affiliation(s)
- Uxia Gurriaran-Rodriguez
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- CIC bioGUNE, Bizkaia Technology Park, Derio, 48160, Spain.
| | - Yves De Repentigny
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada
| | - Rashmi Kothary
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada.
| |
Collapse
|
22
|
Mizuta R, Kanao E, Ukyo K, Kuwada S, Sawada SI, Ishihama Y, Akiyoshi K, Sasaki Y. A Direct Approach for Living Biomembrane Printing on a Nanoparticle. NANO LETTERS 2024. [PMID: 39377259 DOI: 10.1021/acs.nanolett.4c03293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Biomembrane coating technologies have been developed to equip synthetic nanomaterials with natural biointerfaces. We report a one-step method for nondestructively coating the biomembranes of "living" cells onto nanoparticle surfaces. By using simple centrifugation, nanoparticles pass through a concentrated layer of living cells. This process mimics exosome release via endocytosis and exocytosis, preserving the membrane integrity of the source cells. The resulting silica nanoparticles were efficiently coated with membrane components from Raw264.7 cells. Nanoflow-liquid chromatography-tandem mass spectrometry confirmed that the proteins composing the membrane originated from the source cells. Additionally, the biomembrane coating suppressed the phagocytosis of silica nanoparticles by Raw264.7 cells while enhancing their uptake by HeLa cells. Our simple and efficient method for living biomembrane coating holds promise for the development of nanoparticles for medical and pharmaceutical applications.
Collapse
Affiliation(s)
- Ryosuke Mizuta
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Eisuke Kanao
- Laboratory of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Clinical and Analytical Chemistry, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Keigo Ukyo
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shusuke Kuwada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yasushi Ishihama
- Laboratory of Proteomics and Drug Discovery, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Laboratory of Clinical and Analytical Chemistry, National Institute of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka 567-0085, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Yoshihiro Sasaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| |
Collapse
|
23
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2024:e14326. [PMID: 39370572 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpètriêre, Sorbonne Université, INSERM UMR1166, Paris, France
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié-Salpètriêre, Sorbonne Université, INSERM UMR1166, Paris, France
| | - Raul D Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor), University of Sao Paulo Medical School Hospital, Sao Paulo, Brazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Stefano Carugo
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Milan, Italy
- Department of Cardio-Thoracic-Vascular Diseases, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
24
|
Ikezu T, Yang Y, Verderio C, Krämer-Albers EM. Extracellular Vesicle-Mediated Neuron-Glia Communications in the Central Nervous System. J Neurosci 2024; 44:e1170242024. [PMID: 39358029 PMCID: PMC11450539 DOI: 10.1523/jneurosci.1170-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 10/04/2024] Open
Abstract
Communication between neurons and glia significantly influences the development maturation, plasticity, and disease progressions of the nervous system. As a new signaling modality, extracellular vesicles display a diverse role for robust functional regulation of neurons through their protein and nucleic acid cargoes. This review highlights recent breakthroughs in the research of signaling mechanisms between glia and neurons mediated by extracellular vesicles that are important for neural development, axonal maintenance, synaptic functions, and disease progression in the mammalian nervous system. We will discuss the biological roles of extracellular vesicles released from neurons, astroglia, microglia, and oligodendroglia in the nervous system and their implications in neurodegenerative disorders.
Collapse
Affiliation(s)
- Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts 02111
| | - Claudia Verderio
- Department of Biomedical Sciences, CNR Institute of Neuroscience, Università Milano-Bicocca, 20854 Vedano al Lambro (MB), Italy
| | - Eva-Maria Krämer-Albers
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, 55128 Mainz, Rhineland Palatinate, Germany
| |
Collapse
|
25
|
Marjani AA, Nader ND, Aghanejad A. Exosomes as targeted diagnostic biomarkers: Recent studies and trends. Life Sci 2024; 354:122985. [PMID: 39151882 DOI: 10.1016/j.lfs.2024.122985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Different categories of extracellular vesicles (EVs) are identified based on their origin and formation processes. Among these, exosomes (EXOs) originate from endosomal compartments merging with the plasma membrane, forming small lipid vesicles that transport a range of molecular cargo such as nucleic acids, proteins, and lipids. The composition of EXOs varies depending on their cellular source, encompassing various cell types, including neutrophils, dendritic cells, and even tumor cells. Remarkably, EXOs possess inherent stability, low immunogenicity, and compatibility, making them efficient nano vectors for drug delivery. Imaging techniques like bioluminescence, fluorescence, and nuclear imaging are crucial in non-invasively tracking EXOs within living organisms. This process requires the attachment of radionuclides to the EXO's structure without altering its essential characteristics. Real-time imaging of EXOs is vital for their clinical application, and recent advancements in labeling and tracking methodologies provide insights into biodistribution, functionality, and potential pathways for EXO-mediated drug delivery. This review presents updated progress in the diverse applications of EXOs in targeted imaging across various modalities, where they function as contrast agents facilitating tissue visualization and disease tracking. Consequently, EXOs emerge as promising entities in medical diagnostics and imaging.
Collapse
Affiliation(s)
- Aida Abbasi Marjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, United States of America
| | - Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Nuclear Medicine, Faculty of Medicine, Imam Reza General Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Liu S, Zhao H, Jiang T, Wan G, Yan C, Zhang C, Yang X, Chen Z. The Angiogenic Repertoire of Stem Cell Extracellular Vesicles: Demystifying the Molecular Underpinnings for Wound Healing Applications. Stem Cell Rev Rep 2024; 20:1795-1812. [PMID: 39001965 DOI: 10.1007/s12015-024-10762-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2024] [Indexed: 07/15/2024]
Abstract
Stem cells-derived extracellular vesicles (SC-EVs) have emerged as promising therapeutic agents for wound repair, recapitulating the biological effects of parent cells while mitigating immunogenic and tumorigenic risks. These EVs orchestrate wound healing processes, notably through modulating angiogenesis-a critical event in tissue revascularization and regeneration. This study provides a comprehensive overview of the multifaceted mechanisms underpinning the pro-angiogenic capacity of EVs from various stem cell sources within the wound microenvironment. By elucidating the molecular intricacies governing their angiogenic prowess, we aim to unravel the mechanistic repertoire underlying their remarkable potential to accelerate wound healing. Additionally, methods to enhance the angiogenic effects of SC-EVs, current limitations, and future perspectives are highlighted, emphasizing the significant potential of this rapidly advancing field in revolutionizing wound healing strategies.
Collapse
Affiliation(s)
- Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huayuan Zhao
- Department of Urology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gui Wan
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chi Zhang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
Long M, Cheng M. Small extracellular vesicles associated miRNA in myocardial fibrosis. Biochem Biophys Res Commun 2024; 727:150336. [PMID: 38959731 DOI: 10.1016/j.bbrc.2024.150336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/20/2024] [Accepted: 06/29/2024] [Indexed: 07/05/2024]
Abstract
Myocardial fibrosis involves the loss of cardiomyocytes, myocardial fibroblast proliferation, and a reduction in angiogenesis, ultimately leading to heart failure, Given its significant implications, it is crucial to explore novel therapies for myocardial fibrosis. Recently one emerging avenue has been the use of small extracellular vesicles (sEV)-carried miRNA. In this review, we summarize the regulatory role of sEV-carried miRNA in myocardial fibrosis. We explored not only the potential diagnostic value of circulating miRNA as biomarkers for heart disease but also the therapeutic implications of sEV-carried miRNA derived from various cellular sources and applications of modified sEV. This exploration is paramount for researchers striving to develop innovative, cell-free therapies as potential drug candidates for the management of myocardial fibrosis.
Collapse
Affiliation(s)
- Minwen Long
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
28
|
Dong Z, Wang Y, Jin W. Liver cirrhosis: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2024; 5:e721. [PMID: 39290252 PMCID: PMC11406049 DOI: 10.1002/mco2.721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Liver cirrhosis is the end-stage of chronic liver disease, characterized by inflammation, necrosis, advanced fibrosis, and regenerative nodule formation. Long-term inflammation can cause continuous damage to liver tissues and hepatocytes, along with increased vascular tone and portal hypertension. Among them, fibrosis is the necessary stage and essential feature of liver cirrhosis, and effective antifibrosis strategies are commonly considered the key to treating liver cirrhosis. Although different therapeutic strategies aimed at reversing or preventing fibrosis have been developed, the effects have not be more satisfactory. In this review, we discussed abnormal changes in the liver microenvironment that contribute to the progression of liver cirrhosis and highlighted the importance of recent therapeutic strategies, including lifestyle improvement, small molecular agents, traditional Chinese medicine, stem cells, extracellular vesicles, and gut remediation, that regulate liver fibrosis and liver cirrhosis. Meanwhile, therapeutic strategies for nanoparticles are discussed, as are their possible underlying broad application and prospects for ameliorating liver cirrhosis. Finally, we also reviewed the major challenges and opportunities of nanomedicine‒biological environment interactions. We hope this review will provide insights into the pathogenesis and molecular mechanisms of liver cirrhosis, thus facilitating new methods, drug discovery, and better treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Zihe Dong
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| | - Yeying Wang
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| | - Weilin Jin
- The First School of Clinical Medicine Lanzhou University Lanzhou People's Republic of China
- Institute of Cancer Neuroscience Medical Frontier Innovation Research Center The First Hospital of Lanzhou University Lanzhou People's Republic of China
| |
Collapse
|
29
|
Deng SL, Fu Q, Liu Q, Huang FJ, Zhang M, Zhou X. Modulating endothelial cell dynamics in fat grafting: the impact of DLL4 siRNA via adipose stem cell extracellular vesicles. Am J Physiol Cell Physiol 2024; 327:C929-C945. [PMID: 39099421 PMCID: PMC11481985 DOI: 10.1152/ajpcell.00186.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 08/06/2024]
Abstract
In the context of improving the efficacy of autologous fat grafts (AFGs) in reconstructive surgery, this study delineates the novel use of adipose-derived mesenchymal stem cells (ADSCs) and their extracellular vesicles (EVs) as vehicles for delivering delta-like ligand 4 (DLL4) siRNA. The aim was to inhibit DLL4, a gene identified through transcriptome analysis as a critical player in the vascular endothelial cells of AFG tissues, thereby negatively affecting endothelial cell functions and graft survival through the Notch signaling pathway. By engineering ADSC EVs to carry DLL4 siRNA (ADSC EVs-siDLL4), the research demonstrated a marked improvement in endothelial cell proliferation, migration, and lumen formation, and enhanced angiogenesis in vivo, leading to a significant increase in the survival rate of AFGs. This approach presents a significant advancement in the field of tissue engineering and regenerative medicine, offering a potential method to overcome the limitations of current fat grafting techniques.NEW & NOTEWORTHY This study introduces a groundbreaking method for enhancing autologous fat graft survival using adipose-derived stem cell extracellular vesicles (ADSC EVs) to deliver DLL4 siRNA. By targeting the delta-like ligand 4 (DLL4) gene, crucial in endothelial cell dynamics, this innovative approach significantly improves endothelial cell functions and angiogenesis, marking a substantial advancement in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Sen-Lin Deng
- Plastic and Aesthetic Department, People's Hospital of Chongqing Banan District, Chongqing, People's Republic of China
| | - Qiang Fu
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Qing Liu
- Banan District Center for Disease Control and Prevention, Chongqing, People's Republic of China
| | - Fu-Jun Huang
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Miao Zhang
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| | - Xun Zhou
- Department of Dermatology and Cosmetology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, People's Republic of China
| |
Collapse
|
30
|
Li S, Cheng F, Zhang Z, Xu R, Shi H, Yan Y. The role of hepatocyte-derived extracellular vesicles in liver and extrahepatic diseases. Biomed Pharmacother 2024; 180:117502. [PMID: 39357327 DOI: 10.1016/j.biopha.2024.117502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/09/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024] Open
Abstract
Extracellular vesicles (EVs) are vesicle-like bodies with a double membrane structure that are released from the cell membrane or secreted by cells into the extracellular environment. These include exosomes, microvesicles, and apoptotic bodies. There is growing evidence indicating that the composition of liver cell contents changes following injury. The quantity of EVs and the biologically active substances they carry vary depending on the condition of the liver cells. Hepatocytes utilize EVs to modulate the functions of different liver cells and transfer them to distant organs via the systemic circulation, thereby playing a crucial role in intercellular communication. This review provides a concise overview of the research on the effects and potential mechanisms of hepatocyte-derived EVs (Hep-EVs) on liver diseases and extrahepatic diseases under different physiological and pathological conditions. Common liver diseases discussed include non-alcoholic fatty liver disease (NAFLD), viral hepatitis, alcoholic liver disease, drug-induced liver damage, and liver cancer. Given that NAFLD is the most prevalent chronic liver disease globally, this review particularly highlights the use of hepatocyte-derived EVs in NAFLD for disease progression, diagnosis, and treatment.
Collapse
Affiliation(s)
- Shihui Li
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Fang Cheng
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Zhuan Zhang
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ruizi Xu
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Honglei Shi
- Wujin Hospital Affiliated With Jiangsu University, Changzhou Wujin People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213004, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213017, China; Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University (Wujin Clinical College of Xuzhou Medical University), Changzhou 213017, China; Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Changzhou 213017, China.
| |
Collapse
|
31
|
Tati V, Muthukumar V S, Shukla S. Mesenchymal vs. epithelial extracellular vesicles in corneal epithelial repair, apoptosis, and immunomodulation: An in vitro study. Exp Eye Res 2024; 247:110027. [PMID: 39127238 DOI: 10.1016/j.exer.2024.110027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Corneal injuries often lead to epithelial damage, apoptosis, and inflammation which impact visual function. Effective epithelial healing is critical for optimal vision and functioning of the cornea. Mesenchymal stem/stromal cells (MSCs)-derived extracellular vesicles (EVs) present promising avenues for cell-free therapy, however, evaluation of their specific roles in corneal epithelial injury requires further investigations with due consideration to the endogenous human corneal epithelial cell-derived EVs (HCEC-EVs). This study aims to isolate and characterize the EVs from a commonly available human corneal epithelial cell line (HCE-2 [50. B1], ATCC) and evaluate their corneal epithelial repair, anti-apoptotic, and immunomodulatory potential in comparison with human bone marrow mesenchymal stem cell-derived EVs (BM-MSC-EVs) in vitro. Both the BM-MSC- and HCEC-EVs exhibited similar morphology with a diameter <150 nm. However, the yield of EVs from HCECs was higher than that of BM-MSCs. Nanoparticle tracking analysis revealed an average EV size of ∼120 nm, while western blotting confirmed the presence of CD63, CD81, and TSG101, whereas Calnexin could not be detected in the BM-MSC- and HCEC-EVs. The corneal epithelial repair was monitored through in vitro wound healing assay, whereas apoptosis was studied through flow cytometry-based Propidium iodide staining in H2O2-treated cells. IL-1β-stimulated HCECs were treated with BM-MSC- and HCEC-EVs for 24 h and expression of pro- (IL-6 and TNF-α) and anti-inflammatory (IL-10 and TGF-β) cytokines was evaluated through ELISA. Our results, limited to in vitro investigations, suggest that compared with HCEC-EVs, BM-MSC-EVs showed: i) accelerated corneal epithelial healing, ii) enhanced anti-apoptotic potential, and iii) improved anti-inflammatory properties, in cultured HCECs.
Collapse
Affiliation(s)
- Vasudeva Tati
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India
| | - Sai Muthukumar V
- Electron Microscopy Laboratory, Department of Physics, Sri Sathya Sai Institute of Higher Learning, Prasanthi Nilayam, Sri Sathya Sai District, Andhra Pradesh, India
| | - Sachin Shukla
- Prof. Brien Holden Eye Research Centre, Hyderabad Eye Research Foundation, L V Prasad Eye Institute, Hyderabad, 500034, India; Sudhakar and Sreekanth Ravi Stem Cell Biology Laboratory, Centre for Ocular Regeneration, L V Prasad Eye Institute, Hyderabad, 500034, India.
| |
Collapse
|
32
|
Ye J, Li D, Jie Y, Luo H, Zhang W, Qiu C. Exosome-based nanoparticles and cancer immunotherapy. Biomed Pharmacother 2024; 179:117296. [PMID: 39167842 DOI: 10.1016/j.biopha.2024.117296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Over the past decades, cancer immunotherapy has encountered challenges such as immunogenicity, inefficiency, and cytotoxicity. Consequently, exosome-based cancer immunotherapy has gained rapid traction as a promising alternative. Exosomes, a type of extracellular vesicles (EVs) ranging from 50 to 150 nm, are self-originating and exhibit fewer side effects compared to traditional therapies. Exosome-based immunotherapy encompasses three significant areas: cancer vaccination, co-inhibitory checkpoints, and adoptive T-cell therapy. Each of these fields leverages the inherent advantages of exosomes, demonstrating substantial potential for individualized tumor therapy and precision medicine. This review aims to elucidate the reasons behind the promise of exosome-based nanoparticles as cancer therapies by examining their characteristics and summarizing the latest research advancements in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi Province, 330000 China.
| | - Danni Li
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Yiting Jie
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Wenjun Zhang
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Cheng Qiu
- Gastrointestinal Surgery, Pingxiang People's Hospital, Jiangxi Province 330000, China.
| |
Collapse
|
33
|
Hua Y, Jiang P, Dai C, Li M. Extracellular vesicle autoantibodies. J Autoimmun 2024; 149:103322. [PMID: 39341173 DOI: 10.1016/j.jaut.2024.103322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
Autoantibodies are immunoglobulin proteins produced by autoreactive B cells responding to self-antigens. Extracellular vesicles (EVs) are membranous structures released by almost all types of cells and extensively distributed in various biological fluids. Studies have indicated that EVs loaded with self-antigens not only play important roles in antigen presentation and autoantibody production but can also form functional immune complexes with autoantibodies (termed EV autoantibodies). While numerous papers have summarized the production and function of pathogenic autoantibodies in diseases, especially autoimmune diseases, reviews on EV autoantibodies are rare. In this review, we outline the existing knowledge about EVs, autoantibodies, and EV antigens, highlighting the formation of EV autoantibodies and their functions in autoimmune diseases and cancers. In conclusion, EV autoantibodies may be involved in the occurrence of disease(s) and also serve as potential non-invasive markers that could help in the diagnosis and/or prognosis of disease. Additional studies designed to define in more detail the molecular characteristics of EV autoantibodies and their contribution to disease are recommended.
Collapse
Affiliation(s)
- Yan Hua
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Panpan Jiang
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Chunyang Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China
| | - Ming Li
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China; Department of Laboratory Medicine, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China; Core Unit of National Clinical Research Center for Laboratory Medicine of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
34
|
Díaz-Valdez J, Javier-Reyna R, Galindo A, Salazar-Villatoro L, Montaño S, Orozco E. EhVps35, a retromer component, is a key factor in secretion, motility, and tissue invasion by Entamoeba histolytica. Front Cell Infect Microbiol 2024; 14:1467440. [PMID: 39397861 PMCID: PMC11466944 DOI: 10.3389/fcimb.2024.1467440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/30/2024] [Indexed: 10/15/2024] Open
Abstract
In humans and Drosophila melanogaster, the functional convergence of the endosomal sorting complex required for transport (ESCRT) machinery that is in charge of selecting ubiquitinated proteins for sorting into multivesicular bodies, and the retromer, that is the complex responsible for protein recycling to the plasma membrane and Golgi apparatus. ESCRT and retromer complexes are codependent for protein sorting recycling, degradation, and secretion. In this article, we studied the EhVps35 C isoform (referred to as EhVps35), that is the central member of the Entamoeba histolytica retromer, and its relation with the ESCRT machinery during sorting and protein recycling events and their involvement virulence. Our findings revealed that EhVps35 interacts with at least 300 proteins that participate in multiple cellular processes. Laser confocal and transmission electronic microscopy images, as well as secretion assays, revealed that EhVps35 is secreted in vesicles together with EhVps23 and EhADH (both ESCRT machinery proteins). In addition, immunoprecipitation, immunofluorescence, and molecular docking assays revealed the relationship among EhVps35 and other ESCRT machinery proteins. Red blood cell stimulus increased EhVps35 secretion, and the knockdown of the Ehvps35 gene in trophozoites reduced their capacity to migrate and invade tissues. This also impacts the cellular localization of ubiquitin, EhVps23 (ESCRT-I), and EhVps32 (ESCRT-III) proteins, strongly suggesting their functional relationship. Our results, taken together, give evidence that EhVps35 is a key factor in E. histolytica virulence mechanisms and that it, together with the ESCRT machinery components and other regulatory proteins, is involved in vesicle trafficking, secretion, migration, and cell proliferation.
Collapse
Affiliation(s)
- Joselin Díaz-Valdez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Rosario Javier-Reyna
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Ausencio Galindo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Lizbeth Salazar-Villatoro
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| | - Sarita Montaño
- Laboratorio de Bioinformática y Simulación Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Sinaloa, Sinaloa, Mexico
| | - Esther Orozco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (IPN), Ciudad de México, Mexico
| |
Collapse
|
35
|
Huang W, Hua F, Su T, Zhou C, Zhao K, Song D. sEV-mediated lipid droplets transferred from bone marrow adipocytes promote ferroptosis and impair osteoblast function. J Lipid Res 2024; 65:100657. [PMID: 39326787 PMCID: PMC11535364 DOI: 10.1016/j.jlr.2024.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/04/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Osteoporosis is linked to increased bone marrow adipocyte (BMAd) proliferation, which displaces bone-forming cells and alters the local environment. The impact of BMAd lipid droplets on bone health and osteoblast function remains unclear. This study investigates the interplay between BMAd-derived lipid droplets and osteoblast functionality, focusing on ferroptosis pathways. Osteoblast cultures were treated with conditioned media from adipocytes to simulate in vivo conditions. High-throughput mRNA sequencing and Western blot analysis were used to profile changes in gene expression and protein levels related to ferroptosis, oxidative phosphorylation, and osteogenic markers. Cellular assays assessed the direct impact of lipid droplets on osteoblast activity. Results showed that osteoblasts exposed to adipocyte-conditioned media had increased intracellular lipid droplet accumulation, upregulation of ferroptosis-related genes and proteins, and downregulation of oxidative phosphorylation and osteoblast differentiation markers. Treatment with ferroptosis inhibitors reversed the detrimental effects on osteoblasts, indicating the functional relevance of this pathway in osteoporosis. BMAd-derived lipid droplets contribute to osteoblast dysfunction through ferroptosis induction. Inhibiting ferroptosis could preserve osteoblast function and combat osteoporosis-related bone issues, suggesting that modulating lipid metabolism and redox balance in bone cells may be promising for future treatments.
Collapse
Affiliation(s)
- Weibo Huang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Hua
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Su
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenghao Zhou
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangcheng Zhao
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
36
|
Cai N, Zhan X, Zhang Q, Di H, Chen C, Hu Y, Yan X. Red Blood Cell-Derived Small Extracellular Vesicles Inhibit Influenza Virus through Surface-Displayed Sialic Acids. Angew Chem Int Ed Engl 2024:e202413946. [PMID: 39275883 DOI: 10.1002/anie.202413946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/16/2024]
Abstract
Disrupting the conserved multivalent binding of hemagglutinin (HA) on influenza A virus (IAV) to sialic acids (SAs) on the host cell membrane offers a robust strategy to block viral attachment and infection, irrespective of antigenic evolution or drug resistance. In this study, we exploit red blood cell-derived small extracellular vesicles (RBC sEVs) as nanodecoys by harnessing their high abundance of surface-displayed SAs to interact with IAV through multivalent HA-SA interactions. This high-avidity binding inhibits viral adhesion to the cell surface, effectively preventing both attachment and infection in a dose-dependent manner. Notably, enzymatic removal of SAs from RBC sEVs significantly diminishes their anti-IAV efficacy. Our findings indicate that RBC sEVs possess intrinsic anti-IAV properties due to their native multivalent SAs and hold considerable promise as antiviral therapeutics.
Collapse
Affiliation(s)
- Niangui Cai
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaozhen Zhan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Qingyuan Zhang
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Haonan Di
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Chen Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Yunyun Hu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| |
Collapse
|
37
|
Gao M, Sun Q, Zhang R, Shan G, Zhang H, Peng R, Liu M, Sun G, Qiao L, Li Y, He X. Extracellular vesicles-hitchhiking boosts the deep penetration of drugs to amplify anti-tumor efficacy. Biomaterials 2024; 314:122829. [PMID: 39276410 DOI: 10.1016/j.biomaterials.2024.122829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Developing drug delivery systems capable of achieving deep tumor penetration is a challenging task, yet there is a significant demand for such systems in cancer treatment. Hitchhiking on tumor-derived extracellular vesicles (EVs) represents a promising strategy for enhancing drug penetration into tumors. However, the limited drug assembly on EVs restricts its further application. Here, we present a novel approach to efficiently attach antitumor drugs to EVs using an engineered cell membrane-based vector. This vector includes the AS1411 aptamer for tumor-specific targeting, the vesicular stomatitis virus glycoprotein (VSV-G) for tumor cell membrane fusion, and a photosensitizer as the therapeutic agent while ensuring optimal drug encapsulation and stability. Upon injection, photosensitizers are firstly transferred to the tumor cell membrane and subsequently piggybacked onto EVs with the inherent secretion process. By hitchhiking with EVs, photosensitizers can be transferred layer by layer deep into the solid tumors. The results suggest that this EVs-hitchhiking strategy enables photosensitizers to penetrate deeply into tumor tissue, thereby enhancing the efficacy of phototherapy. This study offers broad application prospects for delivering drugs deeply into tumor tissues.
Collapse
Affiliation(s)
- Min Gao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Qiuting Sun
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Ruijie Zhang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Guisong Shan
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Huiru Zhang
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Rui Peng
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Mengyu Liu
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Lei Qiao
- Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Yang Li
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China.
| | - Xiaoyan He
- School of Life Sciences, Anhui Medical University, Hefei, 230032, PR China.
| |
Collapse
|
38
|
Zhang CH, Lu DC, Liu Y, Wang L, Sethi G, Ma Z. The role of extracellular vesicles in pyroptosis-mediated infectious and non-infectious diseases. Int Immunopharmacol 2024; 138:112633. [PMID: 38986299 DOI: 10.1016/j.intimp.2024.112633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/22/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
Pyroptosis, a lytic and pro-inflammatory cell death, is important in various pathophysiological processes. Host- and bacteria-derived extracellular vesicles (EVs), as natural nanocarriers messengers, are versatile mediators of intercellular communication between different types of cells. Recently, emerging research has suggested that EVs exhibit multifaceted roles in disease progression by manipulating pyroptosis. This review focuses on new findings concerning how EVs shape disease progression in infectious and non-infectious diseases by regulating pyroptosis. Understanding the characteristics and activity of EVs-mediated pyroptotic death may conducive to the discovery of novel mechanisms and more efficient therapeutic targets in infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Cai-Hua Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China; Department of Oncology, People's Hospital Affiliated to Chongqing Three Gorges Medical College, Chongqing 404100, China
| | - Ding-Ci Lu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Ying Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600 Singapore; NUS Centre for Cancer Research (N2CR), National University of Singapore, 117599 Singapore.
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China.
| |
Collapse
|
39
|
Notario Manzano R, Chaze T, Rubinstein E, Penard E, Matondo M, Zurzolo C, Brou C. Proteomic landscape of tunneling nanotubes reveals CD9 and CD81 tetraspanins as key regulators. eLife 2024; 13:RP99172. [PMID: 39250349 PMCID: PMC11383530 DOI: 10.7554/elife.99172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
Abstract
Tunneling nanotubes (TNTs) are open actin- and membrane-based channels, connecting remote cells and allowing direct transfer of cellular material (e.g. vesicles, mRNAs, protein aggregates) from the cytoplasm to the cytoplasm. Although they are important especially, in pathological conditions (e.g. cancers, neurodegenerative diseases), their precise composition and their regulation were still poorly described. Here, using a biochemical approach allowing to separate TNTs from cell bodies and from extracellular vesicles and particles (EVPs), we obtained the full composition of TNTs compared to EVPs. We then focused on two major components of our proteomic data, the CD9 and CD81 tetraspanins, and further investigated their specific roles in TNT formation and function. We show that these two tetraspanins have distinct non-redundant functions: CD9 participates in stabilizing TNTs, whereas CD81 expression is required to allow the functional transfer of vesicles in the newly formed TNTs, possibly by regulating docking to or fusion with the opposing cell.
Collapse
Affiliation(s)
- Roberto Notario Manzano
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
- Sorbonne Université, ED394 - Physiologie, Physiopathologie et Thérapeutique, Paris, France
| | - Thibault Chaze
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Eric Rubinstein
- Centre d'Immunologie et des Maladies Infectieuses, Inserm, CNRS, Sorbonne Université, CIMI-Paris, Paris, France
| | - Esthel Penard
- Ultrastructural BioImaging Core Facility (UBI), C2RT, Institut Pasteur, Université Paris Cité, Paris, France
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, CNRS USR 2000, Institut Pasteur, Paris, France
| | - Chiara Zurzolo
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
| | - Christel Brou
- Membrane Traffic and Pathogenesis Unit, Department of Cell Biology and Infection, CNRS 18 UMR 3691, Institut Pasteur, Université Paris Cité, Paris, France
| |
Collapse
|
40
|
Silvestri M, Grazioli E, Duranti G, Sgrò P, Dimauro I. Exploring the Impact of Exercise-Derived Extracellular Vesicles in Cancer Biology. BIOLOGY 2024; 13:701. [PMID: 39336127 PMCID: PMC11429480 DOI: 10.3390/biology13090701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024]
Abstract
Cancer remains a major challenge in medicine, prompting exploration of innovative therapies. Recent studies suggest that exercise-derived extracellular vesicles (EVs) may offer potential anti-cancer benefits. These small, membrane-bound particles, including exosomes, carry bioactive molecules such as proteins and RNA that mediate intercellular communication. Exercise has been shown to increase EV secretion, influencing physiological processes like tissue repair, inflammation, and metabolism. Notably, preclinical studies have demonstrated that exercise-derived EVs can inhibit tumor growth, reduce metastasis, and enhance treatment response. For instance, in a study using animal models, exercise-derived EVs were shown to suppress tumor proliferation in breast and colon cancers. Another study reported that these EVs reduced metastatic potential by decreasing the migration and invasion of cancer cells. Additionally, exercise-induced EVs have been found to enhance the effectiveness of chemotherapy by sensitizing tumor cells to treatment. This review highlights the emerging role of exercise-derived circulating biomolecules, particularly EVs, in cancer biology. It discusses the mechanisms through which EVs impact cancer progression, the challenges in translating preclinical findings to clinical practice, and future research directions. Although research in this area is still limited, current findings suggest that EVs could play a crucial role in spreading molecules that promote better health in cancer patients. Understanding these EV profiles could lead to future therapies, such as exercise mimetics or targeted drugs, to treat cancer.
Collapse
Affiliation(s)
- Monica Silvestri
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
| | - Elisa Grazioli
- Unit of Physical Exercise and Sport Sciences, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
| | - Guglielmo Duranti
- Unit of Biochemistry and Molecular Biology, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
| | - Paolo Sgrò
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy
| |
Collapse
|
41
|
Lawrence SR, Shah KM. Prospects and Current Challenges of Extracellular Vesicle-Based Biomarkers in Cancer. BIOLOGY 2024; 13:694. [PMID: 39336121 PMCID: PMC11428408 DOI: 10.3390/biology13090694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024]
Abstract
Cancer continues to impose a substantial global health burden, particularly among the elderly, where the ongoing global demographic shift towards an ageing population underscores the growing need for early cancer detection. This is essential for enabling personalised cancer care and optimised treatment throughout the disease course to effectively mitigate the increasing societal impact of cancer. Liquid biopsy has emerged as a promising strategy for cancer diagnosis and treatment monitoring, offering a minimally invasive method for the isolation and molecular profiling of circulating tumour-derived components. The expansion of the liquid biopsy approach to include the detection of tumour-derived extracellular vesicles (tdEVs) holds significant therapeutic opportunity. Evidence suggests that tdEVs carry cargo reflecting the contents of their cell-of-origin and are abundant within the blood, exhibiting superior stability compared to non-encapsulated tumour-derived material, such as circulating tumour nucleic acids and proteins. However, despite theoretical promise, several obstacles hinder the translation of extracellular vesicle-based cancer biomarkers into clinical practice. This critical review assesses the current prospects and challenges facing the adoption of tdEV biomarkers in clinical practice, offering insights into future directions and proposing strategies to overcome translational barriers. By addressing these issues, EV-based liquid biopsy approaches could revolutionise cancer diagnostics and management.
Collapse
Affiliation(s)
- Samuel R Lawrence
- Division of Clinical Medicine, School of Medicine & Population Health, The University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| | - Karan M Shah
- Division of Clinical Medicine, School of Medicine & Population Health, The University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK
| |
Collapse
|
42
|
Komori T, Fukuda M. Two roads diverged in a cell: insights from differential exosome regulation in polarized cells. Front Cell Dev Biol 2024; 12:1451988. [PMID: 39286483 PMCID: PMC11402822 DOI: 10.3389/fcell.2024.1451988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Exosomes are extracellular vesicles involved in intercellular signaling, carrying various cargo from microRNAs to metabolites and proteins. They are released by practically all cells and are highly heterogenous due to their origin and content. Several groups of exosomes are known to be involved in various pathological conditions including autoimmune, neurodegenerative, and infectious diseases as well as cancer, and therefore a substantial understanding of their biogenesis and release is crucial. Polarized cells display an array of specific functions originated from differentiated membrane trafficking systems and could lead to hints in untangling the complex process of exosomes. Indeed, recent advances have successfully revealed specific regulation pathways for releasing different subsets of exosomes from different sides of polarized epithelial cells, underscoring the importance of polarized cells in the field. Here we review current evidence on exosome biogenesis and release, especially in polarized cells, highlight the challenges that need to be combatted, and discuss potential applications related to exosomes of polarized-cell origin.
Collapse
Affiliation(s)
- Tadayuki Komori
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
43
|
Suanno G, Genna VG, Maurizi E, Dieh AA, Griffith M, Ferrari G. Cell therapy in the cornea: The emerging role of microenvironment. Prog Retin Eye Res 2024; 102:101275. [PMID: 38797320 DOI: 10.1016/j.preteyeres.2024.101275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The cornea is an ideal testing field for cell therapies. Its highly ordered structure, where specific cell populations are sequestered in different layers, together with its accessibility, has allowed the development of the first stem cell-based therapy approved by the European Medicine Agency. Today, different techniques have been proposed for autologous and allogeneic limbal and non-limbal cell transplantation. Cell replacement has also been attempted in cases of endothelial cell decompensation as it occurs in Fuchs dystrophy: injection of cultivated allogeneic endothelial cells is now in advanced phases of clinical development. Recently, stromal substitutes have been developed with excellent integration capability and transparency. Finally, cell-derived products, such as exosomes obtained from different sources, have been investigated for the treatment of severe corneal diseases with encouraging results. Optimization of the success rate of cell therapies obviously requires high-quality cultured cells/products, but the role of the surrounding microenvironment is equally important to allow engraftment of transplanted cells, to preserve their functions and, ultimately, lead to restoration of tissue integrity and transparency of the cornea.
Collapse
Affiliation(s)
- Giuseppe Suanno
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Eleonora Maurizi
- Centre for Regenerative Medicine ''S. Ferrari'', University of Modena and Reggio Emilia, Modena, Italy
| | - Anas Abu Dieh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - May Griffith
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | - Giulio Ferrari
- Vita-Salute San Raffaele University, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy; Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
44
|
Parikh R, Parikh S, Berzin D, Vaknine H, Ovadia S, Likonen D, Greenberger S, Scope A, Elgavish S, Nevo Y, Plaschkes I, Nizri E, Kobiler O, Maliah A, Zaremba L, Mohan V, Sagi I, Ashery-Padan R, Carmi Y, Luxenburg C, Hoheisel JD, Khaled M, Levesque MP, Levy C. Recycled melanoma-secreted melanosomes regulate tumor-associated macrophage diversification. EMBO J 2024; 43:3553-3586. [PMID: 38719996 PMCID: PMC11377571 DOI: 10.1038/s44318-024-00103-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 02/06/2024] [Accepted: 03/26/2024] [Indexed: 09/07/2024] Open
Abstract
Extracellular vesicles (EVs) are important mediators of communication between cells. Here, we reveal a new mode of intercellular communication by melanosomes, large EVs secreted by melanocytes for melanin transport. Unlike small EVs, which are disintegrated within the receiver cell, melanosomes stay intact within them, gain a unique protein signature, and can then be further transferred to another cell as "second-hand" EVs. We show that melanoma-secreted melanosomes passaged through epidermal keratinocytes or dermal fibroblasts can be further engulfed by resident macrophages. This process leads to macrophage polarization into pro-tumor or pro-immune cell infiltration phenotypes. Melanosomes that are transferred through fibroblasts can carry AKT1, which induces VEGF secretion from macrophages in an mTOR-dependent manner, promoting angiogenesis and metastasis in vivo. In melanoma patients, macrophages that are co-localized with AKT1 are correlated with disease aggressiveness, and immunotherapy non-responders are enriched in macrophages containing melanosome markers. Our findings suggest that interactions mediated by second-hand extracellular vesicles contribute to the formation of the metastatic niche, and that blocking the melanosome cues of macrophage diversification could be helpful in halting melanoma progression.
Collapse
Affiliation(s)
- Roma Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Shivang Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- The Ragon Institute of Mass General, Massachusetts Institute of Technology (MIT), and Harvard, MA 02139, Cambridge, USA
| | - Daniella Berzin
- Institute of Pathology, Sheba Medical Center, Tel Hashomer, 52621, Israel
| | - Hananya Vaknine
- Institute of Pathology, E. Wolfson Medical Center, Holon, 58100, Israel
| | - Shai Ovadia
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Daniela Likonen
- Institute of Pathology, Sheba Medical Center, Tel Hashomer, 52621, Israel
| | | | - Alon Scope
- The Kittner Skin Cancer Screening and Research Institute, Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharona Elgavish
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem and Hadassah Medical Center, Jerusalem, 91120, Israel
| | - Yuval Nevo
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem and Hadassah Medical Center, Jerusalem, 91120, Israel
| | - Inbar Plaschkes
- Info-CORE, Bioinformatics Unit of the I-CORE at the Hebrew University of Jerusalem and Hadassah Medical Center, Jerusalem, 91120, Israel
| | - Eran Nizri
- Department of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, 6423906, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Oren Kobiler
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv Universitygrid.12136.37, Tel Aviv, Israel
| | - Avishai Maliah
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Laureen Zaremba
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Vishnu Mohan
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine and Sagol School of Neurosciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yaron Carmi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Chen Luxenburg
- Cell and Developmental Biology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mehdi Khaled
- INSERM 1279, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Mitchell P Levesque
- Department of Dermatology, University of Zurich, University Hospital Zurich, Wagistrasse 18, CH-8952, Schlieren, Switzerland
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel.
| |
Collapse
|
45
|
Liu C, Sun L, Worden H, Ene J, Zeng OZ, Bhagu J, Grant SC, Bao X, Jung S, Li Y. Profiling biomanufactured extracellular vesicles of human forebrain spheroids in a Vertical-Wheel Bioreactor. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70002. [PMID: 39211409 PMCID: PMC11350274 DOI: 10.1002/jex2.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Extracellular vesicles (EVs) secreted by human brain cells have great potential as cell-free therapies in various diseases, including stroke. However, because of the significant amount of EVs needed in preclinical and clinical trials, EV application is still challenging. Vertical-Wheel Bioreactors (VWBRs) have designed features that allow for scaling up the generation of human forebrain spheroid EVs under low shear stress. In this study, EV secretion by human forebrain spheroids derived from induced pluripotent stem cells as 3D aggregates and on Synthemax II microcarriers in VWBRs were investigated with static aggregate culture as a control. The spheroids were characterized by metabolite and transcriptome analysis. The isolated EVs were characterized by nanoparticle tracking analysis, electron microscopy, and Western blot. The EV cargo was analyzed using proteomics and miRNA sequencing. The in vitro functional assays of an oxygen and glucose-deprived stroke model were conducted. Proof of concept in vivo study was performed, too. Human forebrain spheroid differentiated on microcarriers showed a higher growth rate than 3D aggregates. Microcarrier culture had lower glucose consumption per million cells and lower glycolysis gene expression but higher EV biogenesis genes. EVs from the three culture conditions showed no differences in size, but the yields from high to low were microcarrier cultures, dynamic aggregates, and static aggregates. The cargo is enriched with proteins (proteomics) and miRNAs (miRNA-seq), promoting axon guidance, reducing apoptosis, scavenging reactive oxygen species, and regulating immune responses. Human forebrain spheroid EVs demonstrated the ability to improve recovery in an in vitro stroke model and in vivo. Human forebrain spheroid differentiation in VWBR significantly increased the EV yields (up to 240-750 fold) and EV biogenesis compared to static differentiation due to the dynamic microenvironment and metabolism change. The biomanufactured EVs from VWBRs have exosomal characteristics and more therapeutic cargo and are functional in in vitro assays, which paves the way for future in vivo stroke studies.
Collapse
Affiliation(s)
- Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- Department of Biomedical Sciences, College of MedicineFlorida State UniversityTallahasseeFloridaUSA
| | | | - Justice Ene
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Olivia Z. Zeng
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| | - Jamini Bhagu
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- National High Magnetic Field LaboratoryFlorida State UniversityTallahasseeFloridaUSA
| | - Samuel C. Grant
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
- National High Magnetic Field LaboratoryFlorida State UniversityTallahasseeFloridaUSA
| | - Xiaoping Bao
- Davidson School of Chemical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | | | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU‐FSU College of EngineeringFlorida State UniversityTallahasseeFloridaUSA
| |
Collapse
|
46
|
Yao X, He D, Wei P, Niu Z, Chen H, Li L, Fu P, Wang Y, Lou S, Qian S, Zheng J, Zuo G, Wang K. DNA Nanomaterial-Empowered Surface Engineering of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306852. [PMID: 38041689 DOI: 10.1002/adma.202306852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/30/2023] [Indexed: 12/03/2023]
Abstract
Extracellular vesicles (EVs) are cell-secreted biological nanoparticles that are critical mediators of intercellular communication. They contain diverse bioactive components, which are promising diagnostic biomarkers and therapeutic agents. Their nanosized membrane-bound structures and innate ability to transport functional cargo across major biological barriers make them promising candidates as drug delivery vehicles. However, the complex biology and heterogeneity of EVs pose significant challenges for their controlled and actionable applications in diagnostics and therapeutics. Recently, DNA molecules with high biocompatibility emerge as excellent functional blocks for surface engineering of EVs. The robust Watson-Crick base pairing of DNA molecules and the resulting programmable DNA nanomaterials provide the EV surface with precise structural customization and adjustable physical and chemical properties, creating unprecedented opportunities for EV biomedical applications. This review focuses on the recent advances in the utilization of programmable DNA to engineer EV surfaces. The biology, function, and biomedical applications of EVs are summarized and the state-of-the-art achievements in EV isolation, analysis, and delivery based on DNA nanomaterials are introduced. Finally, the challenges and new frontiers in EV engineering are discussed.
Collapse
Affiliation(s)
- Xuxiang Yao
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Dongdong He
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Pengyao Wei
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Zitong Niu
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
| | - Hao Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Lin Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Pan Fu
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Yiting Wang
- College of Chemistry, Jilin Normal University, Siping, 136000, P. R. China
| | - Saiyun Lou
- Second Clinical Medicine Faculty, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
- Ningbo Second Hospital, Ningbo, 315010, P. R. China
| | - Sihua Qian
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Jianping Zheng
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Guokun Zuo
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, P. R. China
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| | - Kaizhe Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Ningbo Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, 315300, P. R. China
| |
Collapse
|
47
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
48
|
Sies H, Mailloux RJ, Jakob U. Fundamentals of redox regulation in biology. Nat Rev Mol Cell Biol 2024; 25:701-719. [PMID: 38689066 DOI: 10.1038/s41580-024-00730-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Oxidation-reduction (redox) reactions are central to the existence of life. Reactive species of oxygen, nitrogen and sulfur mediate redox control of a wide range of essential cellular processes. Yet, excessive levels of oxidants are associated with ageing and many diseases, including cardiological and neurodegenerative diseases, and cancer. Hence, maintaining the fine-tuned steady-state balance of reactive species production and removal is essential. Here, we discuss new insights into the dynamic maintenance of redox homeostasis (that is, redox homeodynamics) and the principles underlying biological redox organization, termed the 'redox code'. We survey how redox changes result in stress responses by hormesis mechanisms, and how the lifelong cumulative exposure to environmental agents, termed the 'exposome', is communicated to cells through redox signals. Better understanding of the molecular and cellular basis of redox biology will guide novel redox medicine approaches aimed at preventing and treating diseases associated with disturbed redox regulation.
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada.
| | - Ursula Jakob
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
49
|
Luo X, Huang L, Guo Y, Yang Y, Gong P, Ye S, Wang L, Feng Y. Identification of potential candidate miRNAs related to semen quality in seminal plasma extracellular vesicles and sperms of male duck (Anas Platyrhynchos). Poult Sci 2024; 103:103928. [PMID: 39003794 PMCID: PMC11298939 DOI: 10.1016/j.psj.2024.103928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/10/2024] [Accepted: 05/29/2024] [Indexed: 07/16/2024] Open
Abstract
Semen quality is an important indicator that can directly affect fertility. In mammals, miRNAs in seminal plasma extracellular vesicles (SPEVs) and sperms can regulate semen quality. However, relevant regulatory mechanism in duck sperms remains largely unclear. In this study, duck SPEVs were isolated and characterized by transmission electron microscopy (TEM), western blot (WB), and nanoparticle tracking analysis (NTA). To identify the important molecules affecting semen quality, we analysed the miRNA expression in sperms and SPEVs of male ducks in high semen quality group ((DHS, DHSE) and low semen quality group (DLS, DLSE). We identified 94 differentially expressed (DE) miRNAs in the comparison of DHS vs. DLS, and 21 DE miRNAs in DHSE vs. DLSE. Target genes of SPEVs DE miRNAs were enriched in ErbB signaling pathway, glycometabolism, and ECM-receptor interaction pathways (P < 0.05), while the target genes of sperm DE miRNAs were enriched in ribosome (P < 0.05). The miRNA-target-pathway interaction network analyses indicated that 5 DE miRNAs (miR-34c-5p, miR-34b-3p, miR-449a, miR-31-5p, and miR-128-1-5p) targeted the largest number of target genes enriched in MAPK, Wnt and calcium signaling pathways, of which FZD9 and ANAPC11 were involved in multiple biological processes related to sperm functions, indicating their regulatory effects on sperm quality. The comparison of DE miRNAs of SPEVs and sperms found that mir-31-5p and novel-273 could potentially serve as biomarkers for semen quality detection. Our findings enhance the insight into the crucial role of SPEV and sperm miRNAs in regulating semen quality and provide a new perspective for subsequent studies.
Collapse
Affiliation(s)
- Xuliang Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Liming Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yan Guo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China
| | - Yu Yang
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, Wuhan, Hubei 430208, P.R. China
| | - Ping Gong
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, Wuhan, Hubei 430208, P.R. China
| | - Shengqiang Ye
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, Wuhan, Hubei 430208, P.R. China
| | - Lixia Wang
- Wuhan Institute of Animal Husbandry and Veterinary Science, Wuhan Academy of Agricultural Science & Technology, Wuhan, Hubei 430208, P.R. China
| | - Yanping Feng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology and College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, P.R. China.
| |
Collapse
|
50
|
Zhao J, Zhu W, Mao Y, Li X, Ling G, Luo C, Zhang P. Unignored intracellular journey and biomedical applications of extracellular vesicles. Adv Drug Deliv Rev 2024; 212:115388. [PMID: 38969268 DOI: 10.1016/j.addr.2024.115388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/02/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
The intracellular journey of extracellular vesicles (EVs) cannot be ignored in various biological pathological processes. In this review, the biogenesis, biological functions, uptake pathways, intracellular trafficking routes, and biomedical applications of EVs were highlighted. Endosomal escape is a unique mode of EVs release. When vesicles escape from endosomes, they avoid the fate of fusing with lysosomes and being degraded, thus having the opportunity to directly enter the cytoplasm or other organelles. This escape mechanism is crucial for EVs to deliver specific signals or substances. The intracellular trafficking of EVs after endosomal escape is a complex and significant biological process that involves the coordinated work of various cellular structures and molecules. Through the in-depth study of this process, the function and regulatory mechanism of EVs are fully understood, providing new dimensions for future biomedical diagnosis and treatment.
Collapse
Affiliation(s)
- Jiuhong Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Wenjing Zhu
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Yuxuan Mao
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Xiaodan Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Cong Luo
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| |
Collapse
|