77 results on '"David G. Meckes"'
Search Results
2. Phenotypic, metabolic, and biogenesis properties of human stem cell-derived cerebellar spheroids
- Author
-
Timothy Hua, Chang Liu, Sonia Kiran, Kelly Gray, Sunghoon Jung, David G. Meckes, Yan Li, and Qing-Xiang Amy Sang
- Subjects
Medicine ,Science - Abstract
Abstract Human cerebellum consists of high density and complexity of neurons. Thus, it is challenging to differentiate cerebellar-like organoids with similar cellular markers and function to the human brain. Our previous study showed that the combination of retinoic acid (RA), Wingless/integrated (Wnt) activator, and Sonic Hedgehog (SHH) activator promotes cerebellar differentiation from human induced pluripotent stem cells (hiPSCs). This study examined phenotypic, metabolic, and biogenesis in early cerebellar development. Cerebellum spheroids were differentiated from human iPSK3 cells. During day 7–14, RA and Wnt activator CHIR99021 were used and SHH activator purmorphamine (PMR) was added later to promote ventralization. Gene expression for early cerebellar layer markers, metabolism, and extracellular vesicle (EV) biogenesis were characterized. Zinc-induced neurotoxicity was investigated as a proof-of-concept of neurotoxicity study. Flow cytometry results showed that there was no significant difference in NEPH3, PTF1A, OLIG2, and MATH1 protein expression between RCP (RA-CHIR-PMR) versus the control condition. However, the expression of cerebellar genes for the molecular layer (BHLE22), the granule cell layer (GABRB2, PAX6, TMEM266, KCNIP4), the Bergmann glial cells (QK1, DAO), and the Purkinje cell layer (ARHGEF33, KIT, MX1, MYH10, PPP1R17, SCGN) was significantly higher in the RCP condition than the control. The shift in metabolic pathways toward glycolysis was observed for RCP condition. The EV biogenesis marker expression was retained. Mild zinc-induced neurotoxicity may exist when zinc exposure exceeds 1.0 µM. RCP treatment can promote specific cerebellar-like differentiation from hiPSCs indicated by gene expression of early cerebellar markers and regionally enriched genes. The higher cerebellar marker expression is accompanied by the elevated glycolysis with the retained EV biogenesis. This study should advance the understanding of biomarkers during early cerebellar development for cerebellum organoid engineering and neurotoxicity study.
- Published
- 2022
- Full Text
- View/download PDF
3. Correction: Kalvala et al. Cannabidiol-Loaded Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Paclitaxel-Induced Peripheral Neuropathy. Pharmaceutics 2023, 15, 554
- Author
-
Anil Kumar Kalvala, Arvind Bagde, Peggy Arthur, Tanmay Kulkarni, Santanu Bhattacharya, Sunil Surapaneni, Nil Kumar Patel, Ramesh Nimma, Aragaw Gebeyehu, Nagavendra Kommineni, Yan Li, David G. Meckes, Li Sun, Bipika Banjara, Keb Mosley-Kellum, Thanh Cong Dinh, and Mandip Singh
- Subjects
n/a ,Pharmacy and materia medica ,RS1-441 - Abstract
“Yan Li” was not included as an author in the original publication [...]
- Published
- 2023
- Full Text
- View/download PDF
4. Multiplex protein profiling method for extracellular vesicle protein detection
- Author
-
Li Sun and David G. Meckes
- Subjects
Medicine ,Science - Abstract
Abstract Extracellular vesicles (EVs) are small nanometer-sized membrane sacs secreted into biological fluids by all cells. EVs encapsulate proteins, RNAs and metabolites from its origin cell and play important roles in intercellular communication events. Over the past decade, EVs have become a new emerging source for cancer diagnostics. One of the challenges in the study of EVs and there utility as diagnostic biomarkers is the amount of EVs needed for traditional protein analysis methods. Here, we present a new immuno-PCR method that takes advantage of commercially available TotalSeq antibodies containing DNA conjugated oligos to identify immobilized protein analysts using real-time qPCR. Using this method, we demonstrate that multiple EV surface proteins can be profiled simultaneously with high sensitivity and specificity. This approach was also successfully applied to similar protocol using cell and serum samples. We further described the development of a micro-size exclusion chromatography method, where we were able to detect EV surface proteins with as little as 10 μL of human serum when combined with immuno-PCR. Overall, these results show that the immuno-PCR method results in rapid detection of multiple EV markers from small sample volumes in a single tube.
- Published
- 2021
- Full Text
- View/download PDF
5. Engineering extracellular vesicles by three‐dimensional dynamic culture of human mesenchymal stem cells
- Author
-
Xuegang Yuan, Li Sun, Richard Jeske, Dingani Nkosi, Sara B. York, Yuan Liu, Samuel C. Grant, David G. Meckes Jr, and Yan Li
- Subjects
human mesenchymal stem cells ,extracellular vesicles ,exosome ,3D aggregation ,rejuvenation ,immunomodulation ,Cytology ,QH573-671 - Abstract
Abstract Human mesenchymal stem cell (hMSC) derived extracellular vesicles (EVs) have shown therapeutic potential in recent studies. However, the corresponding therapeutic components are largely unknown, and scale‐up production of hMSC EVs is a major challenge for translational applications. In the current study, hMSCs were grown as 3D aggregates under wave motion to promote EV secretion. Results demonstrate that 3D hMSC aggregates promote activation of the endosomal sorting complexes required for transport (ESCRT)‐dependent and ‐independent pathways. mRNA sequencing revealed global transcriptome alterations for 3D hMSC aggregates. Compared to 2D‐hMSC‐EVs, the quantity of 3D‐hMSC‐EVs was enhanced significantly (by 2‐fold), with smaller sizes, higher miR‐21 and miR‐22 expression, and an altered protein cargo (e.g., upregulation of cytokines and anti‐inflammatory factors) uncovered by proteomics analysis, possibly due to altered EV biogenesis. Functionally, 3D‐hMSC‐EVs rejuvenated senescent stem cells and exhibited enhanced immunomodulatory potentials. In summary, this study provides a promising strategy for scalable production of high‐quality EVs from hMSCs with enhanced therapeutic potential.
- Published
- 2022
- Full Text
- View/download PDF
6. Alix and Syntenin-1 direct amyloid precursor protein trafficking into extracellular vesicles
- Author
-
Allaura S. Cone, Stephanie N. Hurwitz, Gloria S. Lee, Xuegang Yuan, Yi Zhou, Yan Li, and David G. Meckes
- Subjects
Extracellular vesicle ,Exosomes ,Alzheimer dementia ,Neurodegeneration ,Multivesicular bodies ,Protein trafficking ,Cytology ,QH573-671 - Abstract
Abstract Background Endosomal trafficking and amyloidogenic cleavage of amyloid precursor protein (APP) is believed to play a role in the neurodegeneration observed in Alzheimer’s disease (AD). Recent evidence has suggested that packaging and secretion of APP and its amyloidogenic cleaved products into small extracellular vesicles (EVs) may facilitate uptake of these neurotoxic factors during disease progression. However, the molecular mechanisms underlying trafficking of APP into EVs are poorly understood. Results In this study, the mechanism and impact of APP trafficking into extracellular vesicles (EVs) were assessed by a series of inducible gene knockdowns. We demonstrate that vesicle-associated proteins Alix and Syntenin-1 are essential for proper subcellular localization and efficient EV secretion of APP via an endosomal sorting complexes required for transport (ESCRT)-independent pathway. The neurotoxic C-terminal fragment (CTFβ) of APP is similarly secreted in association with small vesicles. These mechanisms are conserved in terminally differentiated neuron-like cells. Furthermore, knockdown of Alix and Syntenin-1 alters the subcellular localization of APP, sequestering the precursor protein to endoplasmic reticulum and endolysosomal compartments, respectively. Finally, transfer of small EVs containing mutant APP confers an increase in reactive oxygen species production and neurotoxicity to human induced pluripotent stem cell-derived cortical neurons and naïve primary neurons, an effect that is ameliorated by Alix and Syntenin-1 depletion. Conclusions Altogether these findings elucidate a novel mechanism for understanding the intracellular trafficking of APP and CTFβ into secreted extracellular vesicles, and the resultant potential impact on neurotoxicity in the context of Alzheimer’s disease amyloidopathy.
- Published
- 2020
- Full Text
- View/download PDF
7. Cannabidiol-Loaded Extracellular Vesicles from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Paclitaxel-Induced Peripheral Neuropathy
- Author
-
Anil Kumar Kalvala, Arvind Bagde, Peggy Arthur, Tanmay Kulkarni, Santanu Bhattacharya, Sunil Surapaneni, Nil Kumar Patel, Ramesh Nimma, Aragaw Gebeyehu, Nagavendra Kommineni, Yan Li, David G. Meckes, Li Sun, Bipika Banjara, Keb Mosley-Kellum, Thanh Cong Dinh, and Mandip Singh
- Subjects
extracellular vesicles ,hUCMSCs ,CBD-EVs ,AMPK ,atomic force microscopy ,morphology ,Pharmacy and materia medica ,RS1-441 - Abstract
In cancer patients, chronic paclitaxel (PTX) treatment causes excruciating pain, limiting its use in cancer chemotherapy. The neuroprotective potential of synthetic cannabidiol (CBD) and CBD formulated in extracellular vesicles (CBD-EVs) isolated from human umbilical cord derived mesenchymal stem cells was investigated in C57BL/6J mice with PTX-induced neuropathic pain (PIPN). The particle size of EVs and CBD-EVs, surface roughness, nanomechanical properties, stability, and release studies were all investigated. To develop neuropathy in mice, PTX (8 mg/kg, i.p.) was administered every other day (four doses). In terms of decreasing mechanical and thermal hypersensitivity, CBD-EVs treatment was superior to EVs treatment or CBD treatment alone (p < 0.001). CBD and CBD-EVs significantly reduced mitochondrial dysfunction in dorsal root ganglions and spinal homogenates of PTX-treated animals by modulating the AMPK pathway (p < 0.001). Studies inhibiting the AMPK and 5HT1A receptors found that CBD did not influence the neurobehavioral or mitochondrial function of PIPN. Based on these results, we hypothesize that CBD and CBD-EVs mitigated PIPN by modulating AMPK and mitochondrial function.
- Published
- 2023
- Full Text
- View/download PDF
8. Zika Virus Hijacks Extracellular Vesicle Tetraspanin Pathways for Cell-to-Cell Transmission
- Author
-
Sara B. York, Li Sun, Allaura S. Cone, Leanne C. Duke, Mujeeb R. Cheerathodi, and David G. Meckes
- Subjects
Microbiology ,QR1-502 - Abstract
Zika virus is a reemerging infectious disease that spread rapidly across the Caribbean and South America. Infection of pregnant women during the first trimester has been linked to microcephaly, a neurological condition where babies are born with smaller heads due to abnormal brain development.
- Published
- 2021
- Full Text
- View/download PDF
9. Combined Transcriptomic and Proteomic Profiling to Unravel Osimertinib, CARP-1 Functional Mimetic (CFM 4.17) Formulation and Telmisartan Combo Treatment in NSCLC Tumor Xenografts
- Author
-
Ramesh Nimma, Anil Kumar Kalvala, Nilkumar Patel, Sunil Kumar Surapaneni, Li Sun, Rakesh Singh, Ebony Nottingham, Arvind Bagde, Nagavendra Kommineni, Peggy Arthur, Aakash Nathani, David G. Meckes, and Mandip Singh
- Subjects
epidermal growth factor receptor ,non-small cell lung cancer ,Lamin B2 ,AMPK ,Osimertinib ,RNA seq ,Pharmacy and materia medica ,RS1-441 - Abstract
The epidermal growth factor receptor (EGFR) is highly expressed in many non-small cell lung cancers (NSCLC), necessitating the use of EGFR-tyrosine kinase inhibitors (TKIs) as first-line treatments. Osimertinib (OSM), a third-generation TKI, is routinely used in clinics, but T790M mutations in exon 20 of the EGFR receptor lead to resistance against OSM, necessitating the development of more effective therapeutics. Telmisartan (TLM), OSM, and cell cycle and apoptosis regulatory protein 1 (CARP-1) functional mimetic treatments (CFM4.17) were evaluated in this study against experimental H1975 tumor xenografts to ascertain their anti-cancer effects. Briefly, tumor growth was studied in H1975 xenografts in athymic nude mice, gene and protein expressions were analyzed using next-generation RNA sequencing, proteomics, RT-PCR, and Western blotting. TLM pre-treatment significantly reduced the tumor burden when combined with CFM-4.17 nanoformulation and OSM combination (TLM_CFM-F_OSM) than their respective single treatments or combination of OSM and TLM with CFM 4.17. Data from RNA sequencing and proteomics revealed that TLM_CFM-F_OSM decreased the expression of Lamin B2, STAT3, SOD, NFKB, MMP-1, TGF beta, Sox-2, and PD-L1 proteins while increasing the expression of AMPK proteins, which was also confirmed by RT-PCR, proteomics, and Western blotting. According to our findings, the TLM_CFM-F_OSM combination has a superior anti-cancer effect in the treatment of NSCLC by affecting multiple resistant markers that regulate mitochondrial homeostasis, inflammation, oxidative stress, and apoptosis.
- Published
- 2022
- Full Text
- View/download PDF
10. Epstein-Barr Virus LMP1 Promotes Syntenin-1- and Hrs-Induced Extracellular Vesicle Formation for Its Own Secretion To Increase Cell Proliferation and Migration
- Author
-
Dingani Nkosi, Li Sun, Leanne C. Duke, Nilkumar Patel, Sunil K. Surapaneni, Mandip Singh, and David G. Meckes
- Subjects
exosomes ,LMP1 ,Syntenin-1 ,Epstein-Barr virus ,trafficking ,extracellular vesicles ,Microbiology ,QR1-502 - Abstract
ABSTRACT Extracellular vesicles (EVs) are important mediators of cell-to-cell communication that are involved in both normal processes and pathological conditions. Latent membrane protein 1 (LMP1) is a major viral oncogene that is expressed in most Epstein-Barr virus (EBV)-associated cancers and secreted in EVs. LMP1-modified EVs have the ability to influence recipient cell growth, migration, and differentiation and regulate immune cell function. Despite the significance of LMP1-modified EVs in EBV malignancies, very little is understood about how this protein hijacks the host EV pathway for secretion. Using the biotin identification (BioID) method, we identified LMP1-proximal interacting proteins that are known to play roles in EV formation and protein trafficking. Analysis of the identified LMP1-interacting proteins revealed an enrichment in the ESCRT pathway and associated proteins, including CD63, Syntenin-1, Alix, TSG101, Hrs, and charged multivesicular body proteins (CHMPs). LMP1 transcriptionally upregulated and increased the protein expression of EV biogenesis and secretion genes. Nanoparticle tracking and immunoblot analysis revealed reduced levels of LMP1 EV packaging and of vesicle production following the knockdown of Syntenin-1, Alix, Hrs, and TSG101, with altered endolysosomal trafficking observed when Syntenin-1 and Hrs expression was reduced. Knockdown of specific ESCRT-III subunits (CHMP4B, -5, and -6) impaired LMP1 packaging and secretion into EVs. Finally, we demonstrate that the efficient secretion of LMP1-modified EVs promotes cell attachment, proliferation, and migration and tumor growth. Together, these results begin to shed light on how LMP1 exploits host ESCRT machinery to direct the incorporation of the viral oncoprotein into the EV pathway for secretion to alter the tumor microenvironment. IMPORTANCE LMP1 is a notable viral protein that contributes to the modification of EV content and tumor microenvironment remodeling. LMP1-modified EVs enhance tumor proliferation, migration, and invasion potential and promote radioresistance. Currently, the mechanisms surrounding LMP1 incorporation into the host EV pathways are not well understood. This study revealed that LMP1 utilizes Hrs, Syntenin-1, and specific components of the ESCRT-III complex for release from the cell, enhancement of EV production, and metastatic properties of cancer cells. These findings begin to unravel the mechanism of LMP1 EV trafficking and may provide new targets to control EBV-associated cancers.
- Published
- 2020
- Full Text
- View/download PDF
11. Coordination of Zika Virus Infection and Viroplasm Organization by Microtubules and Microtubule-Organizing Centers
- Author
-
Rebecca A. Buchwalter, Sarah C. Ogden, Sara B. York, Li Sun, Chunfeng Zheng, Christy Hammack, Yichen Cheng, Jieyan V. Chen, Allaura S. Cone, David G. Meckes, Hengli Tang, and Timothy L. Megraw
- Subjects
Zika virus ,ZIKV ,flavivirus ,centrosome ,centriole ,microtubule-organizing center ,Cytology ,QH573-671 - Abstract
Zika virus (ZIKV) became a global health concern in 2016 due to its links to congenital microcephaly and other birth defects. Flaviviruses, including ZIKV, reorganize the endoplasmic reticulum (ER) to form a viroplasm, a compartment where virus particles are assembled. Microtubules (MTs) and microtubule-organizing centers (MTOCs) coordinate structural and trafficking functions in the cell, and MTs also support replication of flaviviruses. Here we investigated the roles of MTs and the cell’s MTOCs on ZIKV viroplasm organization and virus production. We show that a toroidal-shaped viroplasm forms upon ZIKV infection, and MTs are organized at the viroplasm core and surrounding the viroplasm. We show that MTs are necessary for viroplasm organization and impact infectious virus production. In addition, the centrosome and the Golgi MTOC are closely associated with the viroplasm, and the centrosome coordinates the organization of the ZIKV viroplasm toroidal structure. Surprisingly, viroplasm formation and virus production are not significantly impaired when infected cells have no centrosomes and impaired Golgi MTOC, and we show that MTs are anchored to the viroplasm surface in these cells. We propose that the viroplasm is a site of MT organization, and the MTs organized at the viroplasm are sufficient for efficient virus production.
- Published
- 2021
- Full Text
- View/download PDF
12. Epstein-Barr Virus LMP1 Modulates the CD63 Interactome
- Author
-
Mujeeb Cheerathodi, Dingani Nkosi, Allaura S. Cone, Sara B. York, and David G. Meckes
- Subjects
latent membrane protein 1 ,Epstein-Barr virus ,Herpesvirus ,proteomics ,mass spectrometry ,interactions ,Microbiology ,QR1-502 - Abstract
Tetraspanin CD63 is a cluster of cell surface proteins with four transmembrane domains; it is associated with tetraspanin-enriched microdomains and typically localizes to late endosomes and lysosomes. CD63 plays an important role in the cellular trafficking of different proteins, EV cargo sorting, and vesicle formation. We have previously shown that CD63 is important in LMP1 trafficking to EVs, and this also affects LMP1-mediated intracellular signaling including MAPK/ERK, NF-κB, and mTOR activation. Using the BioID method combined with mass spectrometry, we sought to define the broad CD63 interactome and how LMP1 modulates this network of interacting proteins. We identified a total of 1600 total proteins as a network of proximal interacting proteins to CD63. Biological process enrichment analysis revealed significant involvement in signal transduction, cell communication, protein metabolism, and transportation. The CD63-only interactome was enriched in Rab GTPases, SNARE proteins, and sorting nexins, while adding LMP1 into the interactome increased the presence of signaling and ribosomal proteins. Our results showed that LMP1 alters the CD63 interactome, shifting the network of protein enrichment from protein localization and vesicle-mediated transportation to metabolic processes and translation. We also show that LMP1 interacts with mTOR, Nedd4 L, and PP2A, indicating the formation of a multiprotein complex with CD63, thereby potentially regulating LMP1-dependent mTOR signaling. Collectively, the comprehensive analysis of CD63 proximal interacting proteins provides insights into the network of partners required for endocytic trafficking and extracellular vesicle cargo sorting, formation, and secretion.
- Published
- 2021
- Full Text
- View/download PDF
13. Nanoparticle analysis sheds budding insights into genetic drivers of extracellular vesicle biogenesis
- Author
-
Stephanie N. Hurwitz, Meghan M. Conlon, Mark A. Rider, Naomi C. Brownstein, and David G. Meckes
- Subjects
oncosomes ,polyethylene glycol ,endocytosis ,trafficking ,bioinformatics ,tetraspanin ,genome engineering ,Cytology ,QH573-671 - Abstract
Background: Extracellular vesicles (EVs) are important mediators of cell-to-cell communication in healthy and pathological environments. Because EVs are present in a variety of biological fluids and contain molecular signatures of their cell or tissue of origin, they have great diagnostic and prognostic value. The ability of EVs to deliver biologically active proteins, RNAs and lipids to cells has generated interest in developing novel therapeutics. Despite their potential medical use, many of the mechanisms underlying EV biogenesis and secretion remain unknown. Methods: Here, we characterized vesicle secretion across the NCI-60 panel of human cancer cells by nanoparticle tracking analysis. Using CellMiner, the quantity of EVs secreted by each cell line was compared to reference transcriptomics data to identify gene products associated with vesicle secretion. Results: Gene products positively associated with the quantity of exosomal-sized vesicles included vesicular trafficking classes of proteins with Rab GTPase function and sphingolipid metabolism. Positive correlates of larger microvesicle-sized vesicle secretion included gene products involved in cytoskeletal dynamics and exocytosis, as well as Rab GTPase activation. One of the identified targets, CD63, was further evaluated for its role in vesicle secretion. Clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 knockout of the CD63 gene in HEK293 cells resulted in a decrease in small vesicle secretion, suggesting the importance of CD63 in exosome biogenesis. Conclusion: These observations reveal new insights into genes involved in exosome and microvesicle formation, and may provide a means to distinguish EV sub-populations. This study offers a foundation for further exploration of targets involved in EV biogenesis and secretion.
- Published
- 2016
- Full Text
- View/download PDF
14. Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids
- Author
-
Mark Marzano, Julie Bejoy, Mujeeb R. Cheerathodi, Li Sun, Sara B. York, Jing Zhao, Takahisa Kanekiyo, Guojun Bu, David G. Meckes, and Yan Li
- Subjects
induced pluripotent stem cells ,extracellular vesicles ,neural progenitors ,neural degeneration ,cardiac mesoderm ,Cytology ,QH573-671 - Abstract
Extracellular vesicles (EVs) contribute to a variety of signaling processes and the overall physiological and pathological states of stem cells and tissues. Human induced pluripotent stem cells (hiPSCs) have unique characteristics that can mimic embryonic tissue development. There is growing interest in the use of EVs derived from hiPSCs as therapeutics, biomarkers, and drug delivery vehicles. However, little is known about the characteristics of EVs secreted by hiPSCs and paracrine signaling during tissue morphogenesis and lineage specification. Methods: In this study, the physical and biological properties of EVs isolated from hiPSC-derived neural progenitors (ectoderm), hiPSC-derived cardiac cells (mesoderm), and the undifferentiated hiPSCs (healthy iPSK3 and Alzheimer’s-associated SY-UBH lines) were analyzed. Results: Nanoparticle tracking analysis and electron microscopy results indicate that hiPSC-derived EVs have an average size of 100−250 nm. Immunoblot analyses confirmed the enrichment of exosomal markers Alix, CD63, TSG101, and Hsc70 in the purified EV preparations. MicroRNAs including miR-133, miR-155, miR-221, and miR-34a were differently expressed in the EVs isolated from distinct hiPSC lineages. Treatment of cortical spheroids with hiPSC-EVs in vitro resulted in enhanced cell proliferation (indicated by BrdU+ cells) and axonal growth (indicated by β-tubulin III staining). Furthermore, hiPSC-derived EVs exhibited neural protective abilities in Aβ42 oligomer-treated cultures, enhancing cell viability and reducing oxidative stress. Our results demonstrate that the paracrine signaling provided by tissue context-dependent EVs derived from hiPSCs elicit distinct responses to impact the physiological state of cortical spheroids. Overall, this study advances our understanding of cell‒cell communication in the stem cell microenvironment and provides possible therapeutic options for treating neural degeneration.
- Published
- 2019
- Full Text
- View/download PDF
15. Extracellular Vesicle Integrins Distinguish Unique Cancers
- Author
-
Stephanie N. Hurwitz and David G. Meckes
- Subjects
Exosomes ,mass spectrometry ,proteomics ,biomarkers ,cancer ,extracellular vesicles ,microvesicles ,oncosomes ,Microbiology ,QR1-502 - Abstract
The proteomic profile of extracellular vesicles (EVs) has been of increasing interest, particularly in understanding cancer growth, drug resistance, and metastatic behavior. Emerging data suggest that cancer-derived EVs carry an array of oncogenic cargo, including certain integrin proteins that may, in turn, promote cell detachment, migration, and selection of future metastatic sites. We previously reported a large comparison of secreted vesicle protein cargo across sixty diverse human cancer cell lines. Here, we analyze the distinct integrin profiles of these cancer EVs. We further demonstrate the enrichment of integrin receptors in cancer EVs compared to vesicles secreted from benign epithelial cells. The total EV integrin levels, including the quantity of integrins α6, αv, and β1 correlate with tumor stage across a variety of epithelial cancer cells. In particular, integrin α6 also largely reflects breast and ovarian progenitor cell expression, highlighting the utility of this integrin protein as a potential circulating biomarker of certain primary tumors. This study provides preliminary evidence of the value of vesicle-associated integrin proteins in detecting the presence of cancer cells and prediction of tumor stage. Differential expression of integrins across cancer cells and selective packaging of integrins into EVs may contribute to further understanding the development and progression of tumor growth and metastasis across a variety of cancer types.
- Published
- 2019
- Full Text
- View/download PDF
16. Methodological Approaches to Study Extracellular Vesicle miRNAs in Epstein–Barr Virus-Associated Cancers
- Author
-
Li Sun and David G. Meckes
- Subjects
exosomes ,oncosomes ,microvesicles ,microRNA ,extracellular vesicle ,herpesvirus ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
Epstein Barr-virus (EBV) was the first virus identified to be associated with human cancer in 1964 and is found ubiquitously throughout the world’s population. It is now established that EBV contributes to the development and progression of multiple human cancers of both lymphoid and epithelial cell origins. EBV encoded miRNAs play an important role in tumor proliferation, angiogenesis, immune escape, tissue invasion, and metastasis. Recently, EBV miRNAs have been found to be released from infected cancer cells in extracellular vesicles (EVs) and regulate gene expression in neighboring uninfected cells present in the tumor microenvironment and possibly at distal sites. As EVs are abundant in many biological fluids, the viral and cellular miRNAs present within EBV-modified EVs may serve as noninvasion markers for cancer diagnosis and prognosis. In this review, we discuss recent advances in EV isolation and miRNA detection, and provide a complete workflow for EV purification from plasma and deep-sequencing for biomarker discovery.
- Published
- 2018
- Full Text
- View/download PDF
17. Ceramide-dependent trafficking of Epstein-Barr virus LMP1 to small extracellular vesicles
- Author
-
Sara B. York, Stephanie N. Hurwitz, Xia Liu, and David G. Meckes
- Subjects
Virology - Published
- 2023
18. MagPEG: a complete extracellular vesicle isolation/analysis solution
- Author
-
Li Sun, Sara York, Brandon Pate, Yanping Zhang, and David G. Meckes
- Abstract
Current extracellular vesicle (EV) isolation methods depend on large expensive equipment like ultracentrifuges and are laborious and time consuming. There is also currently no method available for high throughput isolation to meet clinical demands. Here, we present a method that combines our previous published ExtraPEG method and magnetic beads. Western blot and nanoparticle tracking analysis (NTA) of the purified EVs revealed higher or equivalent recovery and purity with this method compared to ExtraPEG or size exclusion chromatography (SEC) methods. With this newly developed workflow and automated liquid handling instrument, we have successfully isolated up to 96 EV samples from 5 µL pre-cleared serum in 45 minutes. Moreover, DNA / small RNA / protein purification and profiling steps could be seamlessly integrated into the isolation workflow. To profile EV protein markers, EVs were lysed from the binding step and covalently bound to the surface of the beads. TotalSeq or ELISA antibody can be applied with under a standard protocol. With this extended protocol, researchers can easily complete EV isolation and protein profiling experiment within 8 hours. Taken together, we provide a high throughput method for EV isolation and molecular analyses that may be used for sensitive biomarker detection from biological fluids.
- Published
- 2022
19. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer's disease-like phenotypes in a preclinical mouse model
- Author
-
Xuegang Yuan, Stephanie M. Kenyon, Shawn C. Moseley, Sarah D. Benthem, David G. Meckes, Michael P. Vreones, Li Sun, Leanne C. Duke, Samuel C. Grant, Alina C. Stimmell, David C Hike, Aaron A. Wilber, James Olcese, Allison N. Carrier, Spencer R. Carver, and Allaura S. Cone
- Subjects
Stromal cell ,Amyloid beta ,Medicine (miscellaneous) ,Mice, Transgenic ,Plaque, Amyloid ,exosomes ,Mesenchymal Stem Cell Transplantation ,Hippocampus ,Pathogenesis ,Immunomodulation ,Extracellular Vesicles ,Mice ,Alzheimer Disease ,Medicine ,Animals ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,mesenchymal stem cells ,Amyloid beta-Peptides ,biology ,Glial fibrillary acidic protein ,business.industry ,Mesenchymal stem cell ,Colocalization ,Brain ,Alzheimer's disease ,Microvesicles ,Disease Models, Animal ,Cell culture ,biology.protein ,Cancer research ,business ,microvesicles ,Research Paper - Abstract
Alzheimer's disease (AD) is an irreversible neurodegenerative disorder that affects more than 44 million people worldwide. Despite the high disease burden, there is no effective treatment for people suffering from AD. Mesenchymal stem cells (MSCs) are multipotent stromal cells that have been widely studied due to their therapeutic potential. However, administration of cells has been found to have a multitude of limitations. Recently, extracellular vesicles (EVs) derived from MSCs have been studied as a therapeutic candidate, as they exhibit similar immunoprotective and immunomodulatory abilities as the host human MSCs. Methods: To test the potential therapeutic effects of MSC EVs, human bone-marrow derived MSCs were grown in three-dimensional (3D) cell culture, and small EVs were harvested using differential ultracentrifugation. These small EVs were given to non-transgenic (NT) or 5XFAD (5 familial Alzheimer's disease mutations) mice intranasally (IN) every 4 days for 4 months. The mice were then required to perform a variety of behavioral assays to measure changes in learning and memory. Afterwards, immunohistochemistry was performed on brain slices to measure amyloid beta (Aβ) and glial fibrillary acidic protein (GFAP) levels. Results: The data revealed that 5XFAD mice that received hMSC-EV treatment behaved significantly better in cognitive tests than saline treated 5XFAD mice, with no significant change between EV-treated 5XFAD mice and NT mice. Additionally, we found lower Aβ plaque load in the hippocampus of the EV-treated mice. Finally, less colocalization between GFAP and Aβ plaques was found in the brain of EV-treated mice compared to saline. Conclusions: Taken together, these data suggest that IN administration of MSC-derived EVs can slow down AD pathogenesis.
- Published
- 2021
20. Anticancer and chemosensitization effects of cannabidiol in 2D and 3D cultures of TNBC: involvement of GADD45α, integrin-α5, -β5, -β1, and autophagy
- Author
-
Sunil Kumar Surapaneni, Nilkumar Patel, Li Sun, Nagavendra Kommineni, Anil Kumar Kalvala, Aragaw Gebeyehu, Peggy Arthur, Leanne C. Duke, Ramesh Nimma, David G Meckes, and Mandip Singh
- Subjects
Pharmaceutical Science ,Apoptosis ,Hydrogels ,Triple Negative Breast Neoplasms ,Integrin alpha5 ,Caspase 9 ,Article ,Fibronectins ,Doxorubicin ,Cell Line, Tumor ,Autophagy ,Cannabidiol ,Humans ,Vimentin ,Beclin-1 ,Cell Proliferation - Abstract
To date, promising therapy for triple negative breast cancer (TNBC) remains a serious concern clinically because of poor prognosis, resistance, and recurrence. Herein, anti-cancer potential of synthetic cannabidiol (CBD; Purisys, GA; GMP grade) was explored either alone or as a chemosensitizer followed by post-treatment with doxorubicin (DOX) in TNBC (i.e., MDA-MB-231 and MDA-MB-468) cells. In comparison to 2D cultures, CBD showed greater IC(50) values in 3D (LDP2 hydrogel based) cultures of MDA-MB-231 (6.26-fold higher) and MDA-MB-468 (10.22-fold higher) cells. Next-generation RNA sequencing revealed GADD45A, GADD45G, FASN, LOX, and integrin (i.e., -α5, -β5) genes to be novelly altered by CBD in MDA-MB-231 cells. CIM-16 plate-based migration assay and western blotting disclosed that CBD induces anti-migratory effects in TNBC cells by decreasing fibronectin, vimentin, and integrins-α5, -β5, and -β1. Western blotting, RT-qPCR, and immunocytochemistry revealed that CBD inhibited autophagy (decreased Beclin1, and ATG-5, −7, and −16) of TNBC cells. CBD pre-treatment increased DOX sensitivity in TNBC cells. CBD pre-treatment accompanied by DOX treatment decreased LOX and integrin-α5, and increased caspase 9 protein respectively in MDA-MB-468 cells.
- Published
- 2022
21. EPSTEIN-BARR VIRUS LMP1 ENHANCES LEVELS OF MICROVESICLE-ASSOCIATED PD-L1
- Author
-
Monica Abou Harb, Li Sun, and David G. Meckes
- Abstract
Extracellular vesicles (EVs) circulate throughout the body and carry cargo that can be conferred to proximal or distant cells, making them major delivery vehicles for cellular communication. Epstein-Barr virus (EBV) infected cells release EVs that contain viral proteins such as the major viral oncogene, latent membrane protein 1 (LMP1). LMP1 has been shown to regulate the cellular gene expression of programmed cell death protein 1 ligand (PD-L1). PD-L1, a protein that suppresses the immune system by binding to PD-1, (a receptor found on cytotoxic T cells). PD-L1 has been recently found to be packaged into small EVs contributing to immune evasion of lung cancer cells. Recent studies establish that MVs are shed in very large amounts by tumor cells, and that elevated levels of MVs correlate to disease metastasis and cancers being more aggressive. Here, we demonstrate PD-L1 enrichment in MVs released from nasopharyngeal carcinoma cells and an important function of EBV LMP1 in regulating PD-L1 levels in MVs. These PD-L1+ MVs containing LMP1 likely contribute to the immunosuppressive microenvironment found in EBV-associated cancers.ImportanceAccumulating evidence over the past decade supports that viruses utilize EVs and associated pathways to incorporate viral products to evade eliciting an immune response, while concurrently enabling viral spread or persistence within the host. Considering that viral proteins confer very strong antigenic peptides that will be recognized by T cells, the regulation of the PD-1 pathway by the overexpression of MV-associated PD-L1 may be a strong immune evasion tactic utilized by viruses. The discovery that EBV LMP1 increases PD-L1 microvesicle secretion, identifies a new therapeutic target in immune blockade therapy. We expect that our findings will begin to clarify the mechanism of LMP1-mediated enhanced packaging of PD-L1 into MVs and may produce more specific targets to treat EBV-associated cancers. Consequently, identifying whether a disease is of viral origin through predictive MV biomarkers could further allow for more targeted therapies.
- Published
- 2022
22. Superior Anti-Cancer Activity of Osimertinib in Combination with CARP-1 Functional Mimetic (CFM4.17) and Telmisartan via Lamin B2 and Apoptotic Pathway Against Non-Small Cell Lung Cancer
- Author
-
Ramesh Nimma, Anil Kumar Kalvala, Nil Kumar Patel, Sunil Kumar Surapaneni, Li sun, Rakesh Singh, Ebony Nottingham, Arvind Bagde, Nagavendra Kommineni, Peggy Arthur, Aakash Nathani, David G. Meckes, and Mandip Sachdeva
- Published
- 2022
23. Extracellular Vesicle Collection from Human Stem Cells Grown in Suspension Bioreactors
- Author
-
Xuegang, Yuan, Xingchi, Chen, Changchun, Zeng, David G, Meckes, and Yan, Li
- Subjects
Extracellular Vesicles ,MicroRNAs ,Bioreactors ,Induced Pluripotent Stem Cells ,Humans ,Mesenchymal Stem Cells - Abstract
Extracellular vesicles (EVs) are particles with 100-1000 nm sizes which are secreted by cells for intercellular communication. Meanwhile, studies have found that EVs secreted by human stem cells carry similar characteristics (microRNAs, proteins, metabolites, etc.) from their cell counterpart. Thus, EVs derived from stem cells, especially human induced pluripotent stem cells (hiPSCs) and human mesenchymal stromal/stem cells (hMSCs) are promising candidates for cell-free therapy. However, conventional planar culture is insufficient to produce a large amount of cells or EVs to satisfy clinical requirements. In this chapter, we described feasible approaches to harvest EVs secreted by lineage-specific hiPSCs and undifferentiated hMSCs in suspension bioreactors. Differentiation of hiPSCs to cortical organoids can be performed in suspension bioreactors and the corresponding EVs can be isolated and purified. This scale-up protocol can be applied to a majority of stem cell types with EV collection thus provides useful information for both experimental and biomanufacturing purposes.
- Published
- 2021
24. Cannabidiol Loaded Extracellular Vesicles Sensitize Triple-Negative Breast Cancer to Doxorubicin in both in-vitro and in vivo Models
- Author
-
Sunil Kumar Surapaneni, David G. Meckes, Aragaw Gebeyehu, Nilkumar Patel, Anil Kumar Kalvala, Peggy Arthur, Mandip Singh, Sara B. York, Leanne C. Duke, Arvind Bagde, Nagavendra Kommineni, and Xuegang Yaun
- Subjects
Sonication ,Pharmaceutical Science ,Breast Neoplasms ,Triple Negative Breast Neoplasms ,Pharmacology ,digestive system ,Umbilical cord ,Article ,Extracellular Vesicles ,In vivo ,Cell Line, Tumor ,Zeta potential ,medicine ,Cannabidiol ,Humans ,Doxorubicin ,Chemistry ,Mesenchymal stem cell ,digestive system diseases ,In vitro ,surgical procedures, operative ,medicine.anatomical_structure ,Female ,medicine.drug - Abstract
Extracellular Vesicles (EVs) were isolated from human umbilical cord mesenchymal stem cells (hUCMSCs) and were further encapsulated with cannabidiol (CBD) through sonication method (CBD EVs). CBD EVs displayed an average particle size of 114.1 ± 1.02 nm, zeta potential of -30.26 ± 0.12 mV, entrapment efficiency of 92.3 ± 2.21% and stability for several months at 4 °C. CBD release from the EVs was observed as 50.74 ± 2.44% and 53.99 ± 1.4% at pH 6.8 and pH 7.4, respectively after 48 h. Our in-vitro studies demonstrated that CBD either alone or in EVs form significantly sensitized MDA-MB-231 cells to doxorubicin (DOX) (*P 0.05). Flow cytometry and migration studies revealed that CBD EVs either alone or in combination with DOX induced G1 phase cell cycle arrest and decreased migration of MDA-MB-231 cells, respectively. CBD EVs and DOX combination significantly reduced tumor burden (***P 0.001) in MDA-MB-231 xenograft tumor model. Western blotting and immunocytochemical analysis demonstrated that CBD EVs and DOX combination decreased the expression of proteins involved in inflammation, metastasis and increased the expression of proteins involved in apoptosis. CBD EVs and DOX combination will have profound clinical significance in not only decreasing the side effects but also increasing the therapeutic efficacy of DOX in TNBC.
- Published
- 2021
25. Zika Virus Hijacks Extracellular Vesicle Tetraspanin Pathways for Cell-to-Cell Transmission
- Author
-
Allaura S. Cone, Mujeeb R. Cheerathodi, Sara B. York, Li Sun, Leanne C. Duke, and David G. Meckes
- Subjects
0301 basic medicine ,Microcephaly ,Viral protein ,Hepatitis C virus ,viruses ,exosomes ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Zika virus ,03 medical and health sciences ,Zika ,Tetraspanin ,flavivirus ,CD63 ,medicine ,Secretion ,Molecular Biology ,030102 biochemistry & molecular biology ,Extracellular vesicle ,medicine.disease ,biology.organism_classification ,Editor's Pick ,Virology ,QR1-502 ,Microvesicles ,tetraspanins ,Flavivirus ,030104 developmental biology ,Capsid ,Infectious disease (medical specialty) ,extracellular vesicles ,microvesicles ,Biogenesis ,Research Article - Abstract
Extracellular vesicles (EVs) are membrane-encapsulated structures released by cells which carry signaling factors, proteins and microRNAs that mediate intercellular communication. Accumulating evidence supports an important role of EVs in the progression of neurological conditions and both the spread and pathogenesis of infectious diseases. It has recently been demonstrated that EVs from Hepatitis C virus (HCV) infected individuals and cells contained replicative-competent viral RNA that was capable of infecting hepatocytes. Being a member of the same viral family, it is likely the Zika virus also hijacks EV pathways to package viral components and secrete vesicles that are infectious and potentially less immunogenic. As EVs have been shown to cross blood-brain and placental barriers, it is possible that Zika virus could usurp normal EV biology to gain access to the brain or developing fetus. Here, we demonstrate that Zika virus infected cells secrete distinct EV sub-populations with specific viral protein profiles and infectious genomes. Zika virus infection resulted in the enhanced production of EVs with varying sizes and density compared to those released from non-infected cells. We also show that the EV enriched tetraspanin CD63 regulates the release of EVs, and Zika viral genomes and capsids following infection. Overall, these findings provide evidence for an alternative means of Zika virus transmission and demonstrate the role of EV biogenesis and trafficking proteins in the modulation of Zika infection.ImportanceZika virus is a re-emerging infectious disease that spread rapidly across the Caribbean and South America. Infection of pregnant women during the first trimester has been linked to microcephaly, a neurological condition where babies are born with smaller heads due to abnormal brain development. Babies born with microcephaly can develop convulsions and suffer disabilities as they age. Despite the significance of Zika virus, little is known about how the virus infects the fetus or causes disease. Extracellular vesicles (EVs) are membrane-encapsulated structures released by cells that are present in all biological fluids. EVs carry signaling factors, proteins and microRNAs that mediate intercellular communication. EVs have been shown to be a means by which some viruses can alter cellular environments and cross previously unpassable cellular barriers. Thus gaining a greater understanding of how Zika affects EV cargo may aid in the development of better diagnostics, targeted therapeutics and prophylactic treatments.
- Published
- 2021
26. The Epstein-Barr virus LMP1 interactome: biological implications and therapeutic targets
- Author
-
Mujeeb R. Cheerathodi and David G. Meckes
- Subjects
0301 basic medicine ,Kinase ,Biology ,medicine.disease_cause ,Proteomics ,Interactome ,Epstein–Barr virus ,Virus ,Article ,Cell biology ,03 medical and health sciences ,stomatognathic diseases ,030104 developmental biology ,medicine.anatomical_structure ,Virology ,Tumor Virus ,medicine ,otorhinolaryngologic diseases ,Signal transduction ,Fibroblast - Abstract
The oncogenic potential of Epstein–Barr virus (EBV) is mostly attributed to latent membrane protein 1 (LMP1), which is essential and sufficient for transformation of fibroblast and primary lymphocytes. LMP1 expression results in the activation of multiple signaling cascades like NF-κB and MAP kinases that trigger cell survival and proliferative pathways. LMP1-specific signaling events are mediated through the recruitment of a number of interacting proteins to various signaling domains. Based on these properties, LMP1 is an attractive target to develop effective therapeutics to treat EBV-related malignancies. In this review, we focus on LMP1-interacting proteins, associated signaling events and potential targets that could be exploited for therapeutic strategies.
- Published
- 2021
27. Extracellular Vesicles in Epstein-Barr Virus Pathogenesis
- Author
-
David G. Meckes, Allaura S. Cone, and Sara B. York
- Subjects
Microbiology (medical) ,Viral protein ,Viral pathogenesis ,RNA ,Biology ,medicine.disease_cause ,Epstein–Barr virus ,Article ,Virus ,Microvesicles ,Pathogenesis ,Infectious Diseases ,hemic and lymphatic diseases ,microRNA ,medicine ,Cancer research - Abstract
PURPOSE OF REVIEW: Epstein-Barr virus (EBV) is a known determinant for numerous malignancies and may contribute to autoimmune diseases. The underlining mechanisms behind EBV pathologies is not completely understood. Recently, extracellular vesicles (EVs) released from infected cells have been found to produce profound effects on cellular microenvironments. Therefore, in this review we sought to critically evaluate the roles of EVs in EBV pathogenesis and assess their potential therapeutic and diagnostic utility. RECENT FINDINGS: EBV-altered EVs are capable of activating signaling cascades and phenotypic changes in recipient cells through the transfer of viral proteins and RNAs. Moreover, several EV-associated microRNAs have encouraging prognostic or diagnostic potential in EBV-associated cancers. SUMMARY: Current evidence suggests that EBV-modified EVs affect viral pathogenesis and cancer progression. However, further research is needed to investigate the direct role of both viral and host products on recipient cells and the mechanisms driving viral protein and RNA EV packaging and content modification.
- Published
- 2019
28. Epstein-Barr Virus LMP1 Modulates the CD63 Interactome
- Author
-
Sara B. York, Allaura S. Cone, David G. Meckes, Mujeeb R. Cheerathodi, and Dingani Nkosi
- Subjects
0301 basic medicine ,Cell signaling ,autophagy ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Endosome ,Sorting Nexins ,Endocytic cycle ,lcsh:QR1-502 ,exosomes ,Interactome ,lcsh:Microbiology ,Article ,Viral Matrix Proteins ,03 medical and health sciences ,Extracellular Vesicles ,0302 clinical medicine ,proteomics ,Tetraspanin ,Virology ,CD63 ,biochemistry ,Epstein-Barr virus ,Humans ,mass spectrometry ,latent membrane protein 1 ,Chemistry ,Tetraspanin 30 ,Herpesvirus ,Extracellular vesicle ,interactions ,Cell biology ,Protein Transport ,030104 developmental biology ,Infectious Diseases ,tetraspanin ,HEK293 Cells ,030220 oncology & carcinogenesis ,embryonic structures ,mTOR ,Rab ,signaling ,Signal Transduction - Abstract
Tetraspanin CD63 is a cluster of cell surface proteins with four transmembrane domains, it is associated with tetraspanin-enriched microdomains and typically localizes to late endosomes and lysosomes. CD63 plays an important role in the cellular trafficking of different proteins, EV cargo sorting, and vesicle formation. We have previously shown that CD63 is important in LMP1 trafficking to EVs, and this also affects LMP1-mediated intracellular signaling including MAPK/ERK, NF-κB, and mTOR activation. Using the BioID method combined with mass spectrometry, we sought to define the broad CD63 interactome and how LMP1 modulates this network of interacting proteins. We identified a total of 1600 total proteins as a network of proximal interacting proteins to CD63. Biological process enrichment analysis revealed significant involvement in signal transduction, cell communication, protein metabolism, and transportation. The CD63-only interactome was enriched in Rab GTPases, SNARE proteins, and sorting nexins, while adding LMP1 into the interactome increased the presence of signaling and ribosomal proteins. Our results showed that LMP1 alters the CD63 interactome, shifting the network of protein enrichment from protein localization and vesicle-mediated transportation to metabolic processes and translation. We also show that LMP1 interacts with mTOR, Nedd4 L, and PP2A, indicating the formation of a multiprotein complex with CD63, thereby potentially regulating LMP1-dependent mTOR signaling. Collectively, the comprehensive analysis of CD63 proximal interacting proteins provides insights into the network of partners required for endocytic trafficking and extracellular vesicle cargo sorting, formation, and secretion.
- Published
- 2021
29. Biogenesis of Extracellular Vesicles Produced from Human-Stem-Cell-Derived Cortical Spheroids Exposed to Iron Oxides
- Author
-
Shannon Helsper, Samuel C. Grant, Yan Li, Mark Marzano, Mayassa J. Bou-Dargham, Sara B. York, Allaura S. Cone, David G. Meckes, and Qing-Xiang Amy Sang
- Subjects
Iron ,0206 medical engineering ,Induced Pluripotent Stem Cells ,Biomedical Engineering ,Iron oxide ,Spheroid ,Oxides ,02 engineering and technology ,Membrane budding ,021001 nanoscience & nanotechnology ,020601 biomedical engineering ,Exosome ,Ferric Compounds ,Article ,Biomaterials ,chemistry.chemical_compound ,Extracellular Vesicles ,chemistry ,Biophysics ,Humans ,MTT assay ,Stem cell ,Progenitor cell ,0210 nano-technology ,Iron oxide nanoparticles - Abstract
Stem-cell-derived extracellular vesicles (EVs) are promising tools for therapeutic delivery and imaging in the medical research fields. EVs that arise from endosomal compartments or plasma membrane budding consist of exosomes and microvesicles, which range between 30 and 200 nm and 100–1000 nm, respectively. Iron oxide nanoparticles can be used to label stem cells or possibly EVs for magnetic resonance imaging. This could be a novel way to visualize areas in the body that are affected by neurological disorders such as stroke. Human induced pluripotent stem cells (iPSK3 cells) were plated on low-attachment plates and treated with SB431542 and LDN193189 during the first week for the induction of cortical spheroid formation and grown with fibroblast growth factor 2 and cyclopamine during the second week for the neural progenitor cell (iNPC) differentiation. iNPCs were then grown on attachment plates and treated with iron oxide (Fe(3)O(4)) nanoparticles at different sizes (8, 15, and 30 nm in diameter) and concentrations (0.1, 10, and 100 μM). The spheroids and media collected from these cultures were used for iron oxide detection as well as EV isolation and characterizations, respectively. MTT assay demonstrated that the increased size and concentration of the iron oxide nanoparticles had little effect on the metabolic activity of iNPCs. In addition, the Live/Dead assay showed high viability in all the nanoparticle treated groups and the untreated control. The EVs isolated from these culture groups were analyzed and displayed similar or higher EV counts compared with control. The observed EV size averaged 200–250 nm, and electron microscopy revealed the expected exosome morphology for EVs from all groups. RT-PCR analysis of EV biogenesis markers (CD63, CD81, Alix, TSG101, Syntenin1, ADAM10, RAB27b, and Syndecan) showed differential expression between the iron-oxide-treated cultures and nontreated cultures, as well as between adherent and nonadherent 3D cultures. Iron oxide nanoparticles were detected inside the cortical spheroid cells but not EVs by MRI. The addition of iron oxide nanoparticles does not induce significant cytotoxic effects to cortical spheroids. In addition,, nanoparticles may stimulate the biogenesis of EVs when added to cortical spheroids in vitro.
- Published
- 2021
30. Extracellular Vesicle Collection from Human Stem Cells Grown in Suspension Bioreactors
- Author
-
Changchun Zeng, Xuegang Yuan, Xingchi Chen, Yan Li, and David G. Meckes
- Subjects
Differential centrifugation ,Stromal cell ,medicine.anatomical_structure ,Chemistry ,Cell ,Mesenchymal stem cell ,Organoid ,medicine ,Biomanufacturing ,Extracellular vesicle ,Stem cell ,Cell biology - Abstract
Extracellular vesicles (EVs) are particles with 100-1000 nm sizes which are secreted by cells for intercellular communication. Meanwhile, studies have found that EVs secreted by human stem cells carry similar characteristics (microRNAs, proteins, metabolites, etc.) from their cell counterpart. Thus, EVs derived from stem cells, especially human induced pluripotent stem cells (hiPSCs) and human mesenchymal stromal/stem cells (hMSCs) are promising candidates for cell-free therapy. However, conventional planar culture is insufficient to produce a large amount of cells or EVs to satisfy clinical requirements. In this chapter, we described feasible approaches to harvest EVs secreted by lineage-specific hiPSCs and undifferentiated hMSCs in suspension bioreactors. Differentiation of hiPSCs to cortical organoids can be performed in suspension bioreactors and the corresponding EVs can be isolated and purified. This scale-up protocol can be applied to a majority of stem cell types with EV collection thus provides useful information for both experimental and biomanufacturing purposes.
- Published
- 2021
31. Micro size exclusion chromatography combined with a multiplex protein profiling method for extracellular vesicle protein detection from small sample volumes
- Author
-
Li Sun and David G. Meckes
- Subjects
Protein profiling ,Chromatography ,Chemistry ,Size-exclusion chromatography ,Small sample ,Multiplex ,Extracellular vesicle ,Protein detection - Abstract
Extracellular vesicles (EVs) are small nanometer-sized membrane sacs secreted into biological fluids by all cells. EVs encapsulate proteins, RNAs and metabolites from its origin cell and play important roles in intercellular communication events. Over the past decade, EVs have become a new emerging source for cancer diagnostics. One of the challenges in the study of EVs and there utility as diagnostic biomarkers is the amount of EVs needed for traditional protein analysis methods. Here, we present a new immuno-PCR method that takes advantage of commercially available TotalSeq™ antibodies containing DNA conjugated oligos to identify immobilized protein analysts using real-time qPCR. Using this method, we demonstrate that multiple EV surface proteins can be profiled simultaneously with high sensitivity and specificity. This approach was also successfully applied to similar protocol using live cell and serum samples. We further described the development of a micro-size exclusion chromatography method, where we were able to detect EV surface proteins with as little as 10 μL of human serum when combined with immuno-PCR. Overall, these results show that the immuno-PCR method results in rapid detection of multiple EV markers from small sample volumes in a single tube.
- Published
- 2020
32. Epstein-Barr virus LMP1 manipulates the content and functions of extracellular vesicles to enhance metastatic potential of recipient cells
- Author
-
Leanne C. Duke, Dingani Nkosi, David G. Meckes, and Li Sun
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cell ,Gene Expression ,Biochemistry ,Metastasis ,Extracellular matrix ,0302 clinical medicine ,Cell Movement ,Basic Cancer Research ,Medicine and Health Sciences ,Tumor Microenvironment ,Biology (General) ,0303 health sciences ,Nasopharyngeal Carcinoma ,Chemistry ,Cell biology ,Nucleic acids ,Crosstalk (biology) ,Cell Motility ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Signal transduction ,Cellular Structures and Organelles ,Research Article ,Signal Transduction ,QH301-705.5 ,Immunology ,Cell Migration ,Microbiology ,Viral Matrix Proteins ,03 medical and health sciences ,Extracellular Vesicles ,Virology ,Cell Line, Tumor ,medicine ,Genetics ,Cell Adhesion ,Humans ,Neoplasm Invasiveness ,Vesicles ,Cell adhesion ,Non-coding RNA ,Molecular Biology ,Protein kinase B ,030304 developmental biology ,Cell Proliferation ,Tumor microenvironment ,Natural antisense transcripts ,Cell growth ,Carcinoma ,Biology and Life Sciences ,Cancers and Neoplasms ,Nasopharyngeal Neoplasms ,Cell Biology ,RC581-607 ,Gene regulation ,MicroRNAs ,RNA ,Parasitology ,Immunologic diseases. Allergy ,Developmental Biology - Abstract
Extracellular vesicles (EV) mediate intercellular communication events and alterations in normal vesicle content contribute to function and disease initiation or progression. The ability to package a variety of cargo and transmit molecular information between cells renders EVs important mediators of cell-to-cell crosstalk. Latent membrane protein 1 (LMP1) is a chief viral oncoprotein expressed in most Epstein-Barr virus (EBV)-associated cancers and is released from cells at high levels in EVs. LMP1 containing EVs have been demonstrated to promote cell growth, migration, differentiation, and regulate immune cell function. Despite these significant changes in recipient cells induced by LMP1 modified EVs, the mechanism how this viral oncogene modulates the recipient cells towards these phenotypes is not well understood. We hypothesize that LMP1 alters EV content and following uptake of the LMP1-modified EVs by the recipient cells results in the activation of cell signaling pathways and increased gene expression which modulates the biological properties of recipient cell towards a new phenotype. Our results show that LMP1 expression alters the EV protein and microRNA content packaged into EVs. The LMP1-modified EVs also enhance recipient cell adhesion, proliferation, migration, invasion concomitant with the activation of ERK, AKT, and NF-κB signaling pathways. The LMP1 containing EVs induced transcriptome reprogramming in the recipient cells by altering gene expression of different targets including cadherins, matrix metalloproteinases 9 (MMP9), MMP2 and integrin-α5 which contribute to extracellular matrix (ECM) remodeling. Altogether, our data demonstrate the mechanism in which LMP1-modified EVs reshape the tumor microenvironment by increasing gene expression of ECM interaction proteins., Author summary Extracellular vesicles (EV) facilitate cell-to-cell crosstalk due to their capability to sort and transfer various cargoes. Multiple studies have shown changes in EV content and cargo affect their functions and contribute to pathological conditions such as cancer. EVs represent a mechanism through which cancer cells modify their microenvironment to enhance growth and metastasis. This study showed that LMP1, an EBV major oncoprotein which is released in EVs alters the EV content and cargo leading enhanced cell attachment, proliferation, migration and invasion. LMP1 modified EVs mediate the transfer of signaling molecules to recipient cells where they induce NF-κB, AKT and MAPK/ERK signaling pathways leading to alteration in gene expression especially those involved in ECM interaction. LMP1 containing EVs modify the microenvironment by upregulating cadherins, fibronectin, integrin-α5, MMP9 and MMP2 to promote a tumor permissive niche leading tumorigenesis or metastasis.
- Published
- 2020
33. Alix and Syntenin-1 direct amyloid precursor protein and amyloid beta trafficking into extracellular vesicles
- Author
-
Stephanie N. Hurwitz, Glorida S. Lee, Yi Zhou, Allaura S. Cone, Xuegang Yuan, David G. Meckes, and Yan Li
- Subjects
biology ,Amyloid beta ,Chemistry ,mental disorders ,Amyloid precursor protein ,biology.protein ,Syntenin-1 ,Extracellular vesicles ,Cell biology - Abstract
Background: Endosomal trafficking and amyloidogenic cleavage of amyloid precursor protein (APP) is believed to play a role in the neurodegeneration observed in Alzheimer’s disease (AD). Recent evidence has suggested that packaging and secretion of APP and amyloid beta into small extracellular vesicles (EVs) may facilitate uptake of these neurotoxic factors during disease progression. However, the molecular mechanisms underlying trafficking of APP into EVs are poorly understood. Results: In this study, the mechanism and impact of amyloid precursor protein (APP) trafficking into extracellular vesicles (EVs) were assessed by a series of inducible gene knockdowns. We demonstrate that vesicle-associated proteins Alix and Syntenin-1 are essential for proper subcellular localization and efficient EV secretion of APP via an endosomal sorting complexes required for transport (ESCRT)-independent pathway. The neurotoxic metabolite amyloid beta (Aβ) is similarly secreted in association with small vesicles. These mechanisms are conserved in terminally differentiated neuron-like cells. Furthermore, knockdown of Alix and Syntenin-1 alters the subcellular localization of APP, sequestering the precursor protein to endoplasmic reticulum and endolysosomal compartments, respectively. Finally, transfer of small EVs containing APP confers an increase in reactive oxygen species production and neurotoxicity to human induced pluripotent stem cell-derived cortical neurons and naïve primary neurons, an effect that is ameliorated by Alix and Syntenin-1 depletion. Conclusions: Altogether these findings elucidate a novel mechanism for understanding the intracellular trafficking of APP and Aβ into secreted extracellular vesicles, and the resultant potential impact on neurotoxicity in the context of Alzheimer’s disease amyloidopathy.
- Published
- 2020
34. An optimized method for enrichment of whole brain-derived extracellular vesicles reveals insight into neurodegenerative processes in a mouse model of Alzheimer’s disease
- Author
-
James Olcese, Li Sun, Kalonji Y. Cole, Charles R. Ford, David G. Meckes, and Stephanie N. Hurwitz
- Subjects
Male ,0301 basic medicine ,Transgene ,Cell ,Mice, Transgenic ,tau Proteins ,Proteomics ,Article ,Amyloid beta-Protein Precursor ,Extracellular Vesicles ,Mice ,03 medical and health sciences ,Microscopy, Electron, Transmission ,Alzheimer Disease ,Presenilin-1 ,medicine ,Animals ,Amyloid beta-Peptides ,Chemistry ,General Neuroscience ,Vesicle ,Neurodegeneration ,Age Factors ,Brain ,Neurodegenerative Diseases ,Extracellular vesicle ,medicine.disease ,Peptide Fragments ,Microvesicles ,Transport protein ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Mutation ,Female - Abstract
Background Alzheimer’s disease (AD) is the major cause of dementia that has increased dramatically in prevalence over the past several decades. Yet many questions still surround the etiology of AD. Recently, extracellular vesicles (EVs) that transport protein, lipid, and nucleic acids from cell to cell have been implicated in the clearance and propagation of misfolded proteins. Investigation of EVs in AD progression, and their potential diagnostic utility may contribute to understanding and treating AD. However, the challenges of isolating brain-derived EVs have in part hindered these studies. New method Here, we provide an optimized method for the enrichment of brain-derived EVs by iodixanol floatation density gradient for mass spectrometry analysis. Results We demonstrate the isolation of these vesicles and the enrichment of EV proteins compared to sedimentation gradient isolation of vesicles. Moreover, comparative proteomic analysis of brain-derived EVs from healthy and AD mouse brains revealed differences in vesicular content including proteins involved in aging, immune response, and oxidation-reduction maintenance. These changes provide insight into AD-associated neurodegeneration and potential biomarkers of AD. Comparison with existing methods: Recent techniques have used sedimentation sucrose gradients to isolate EVs from brain tissue. However, here we demonstrate the advantages of floatation iodixanol density gradient isolation of small EVs, and provide evidence of EV enrichment by electron microscopy, immunoblot analysis, and quantitative mass spectrometry. Conclusions Together these findings offer a rigorous technique for enriching whole tissue-derived EVs for downstream analyses, and application of this approach to uncovering molecular changes in AD progression and other neurological conditions.
- Published
- 2018
35. Coordination of Zika Virus Infection and Viroplasm Organization by Microtubules and Microtubule-Organizing Centers
- Author
-
Hengli Tang, Christy Hammack, Sara B. York, Yichen Cheng, Rebecca A Buchwalter, Jieyan V. Chen, Li Sun, David G. Meckes, Chunfeng Zheng, Sarah C. Ogden, Allaura S. Cone, and Timothy L. Megraw
- Subjects
viroplasm ,Centriole ,QH301-705.5 ,Golgi Apparatus ,Endoplasmic Reticulum ,Microtubules ,Article ,Zika virus ,Cell Line ,symbols.namesake ,flavivirus ,Microtubule ,centriole ,Humans ,Viroplasm ,ZIKV ,centrosome ,microtubule-organizing center ,MTOC ,microtubule ,Biology (General) ,Centrosome ,biology ,Zika Virus Infection ,Virion ,Microtubule organizing center ,General Medicine ,Golgi apparatus ,biology.organism_classification ,Cell biology ,Flavivirus ,symbols ,Viral Replication Compartments - Abstract
Zika virus (ZIKV) became a global health concern in 2016 due to its links to congenital microcephaly and other birth defects. Flaviviruses, including ZIKV, reorganize the endoplasmic reticulum (ER) to form a viroplasm, a compartment where virus particles are assembled. Microtubules (MTs) and microtubule-organizing centers (MTOCs) coordinate structural and trafficking functions in the cell, and MTs also support replication of flaviviruses. Here we investigated the roles of MTs and the cell’s MTOCs on ZIKV viroplasm organization and virus production. We show that a toroidal-shaped viroplasm forms upon ZIKV infection, and MTs are organized at the viroplasm core and surrounding the viroplasm. We show that MTs are necessary for viroplasm organization and impact infectious virus production. In addition, the centrosome and the Golgi MTOC are closely associated with the viroplasm, and the centrosome coordinates the organization of the ZIKV viroplasm toroidal structure. Surprisingly, viroplasm formation and virus production are not significantly impaired when infected cells have no centrosomes and impaired Golgi MTOC, and we show that MTs are anchored to the viroplasm surface in these cells. We propose that the viroplasm is a site of MT organization, and the MTs organized at the viroplasm are sufficient for efficient virus production.
- Published
- 2021
36. BioID Combined with Mass Spectrometry to Study Herpesvirus Protein-Protein Interaction Networks
- Author
-
Mujeeb R, Cheerathodi and David G, Meckes
- Subjects
Viral Proteins ,Humans ,Herpesviridae Infections ,Protein Interaction Maps ,Herpesviridae ,Mass Spectrometry ,Cell Line ,Chromatography, Liquid - Abstract
Herpes viruses are important human pathogens that cause a wide range of diseases from skin lesions to malignancies. Protein interactions drive many cellular events and mediate a number of biochemical pathways leading to different physiological outcomes. Protein interactions between viral proteins and host proteins play significant roles in viral entry, replication and suppression of host-immune responses. Therefore, the study of virus-host interactions promises significant advancement in designing therapeutics to control infection and disease. Various approaches are employed in the field to study and identify protein interactions that combine affinity purification along with different detection methods. Advancements in protein purification and high-throughput detection methods have resulted in an unprecedented level of discovery. Here we detail the use of proximity dependent biotinylation (BioID) as a means of affinity purification coupled with the use of LC-MS/MS for the detection and identification of protein-protein interaction networks.
- Published
- 2019
37. BioID Combined with Mass Spectrometry to Study Herpesvirus Protein–Protein Interaction Networks
- Author
-
David G. Meckes and Mujeeb R. Cheerathodi
- Subjects
0303 health sciences ,030302 biochemistry & molecular biology ,Human pathogen ,Computational biology ,Biology ,Proteomics ,Protein protein interaction network ,Protein–protein interaction ,03 medical and health sciences ,Affinity chromatography ,Viral entry ,Biotinylation ,Protein purification ,030304 developmental biology - Abstract
Herpes viruses are important human pathogens that cause a wide range of diseases from skin lesions to malignancies. Protein interactions drive many cellular events and mediate a number of biochemical pathways leading to different physiological outcomes. Protein interactions between viral proteins and host proteins play significant roles in viral entry, replication and suppression of host-immune responses. Therefore, the study of virus-host interactions promises significant advancement in designing therapeutics to control infection and disease. Various approaches are employed in the field to study and identify protein interactions that combine affinity purification along with different detection methods. Advancements in protein purification and high-throughput detection methods have resulted in an unprecedented level of discovery. Here we detail the use of proximity dependent biotinylation (BioID) as a means of affinity purification coupled with the use of LC-MS/MS for the detection and identification of protein-protein interaction networks.
- Published
- 2019
38. Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids
- Author
-
Yan Li, Julie Bejoy, Li Sun, Guojun Bu, Jing Zhao, Mujeeb R. Cheerathodi, David G. Meckes, Takahisa Kanekiyo, Mark Marzano, and Sara B. York
- Subjects
Mesoderm ,Cell Survival ,induced pluripotent stem cells ,Morphogenesis ,Neural degeneration ,neural progenitors ,Biology ,Article ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,Spheroids, Cellular ,Ectoderm ,Paracrine Communication ,medicine ,Humans ,neural degeneration ,Viability assay ,Progenitor cell ,Particle Size ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Cells, Cultured ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,Cell Differentiation ,General Medicine ,3. Good health ,Cell biology ,MicroRNAs ,medicine.anatomical_structure ,lcsh:Biology (General) ,cardiac mesoderm ,Nanoparticles ,Stem cell ,extracellular vesicles ,030217 neurology & neurosurgery ,Biomarkers - Abstract
Extracellular vesicles (EVs) contribute to a variety of signaling processes and the overall physiological and pathological states of stem cells and tissues. Human induced pluripotent stem cells (hiPSCs) have unique characteristics that can mimic embryonic tissue development. There is growing interest in the use of EVs derived from hiPSCs as therapeutics, biomarkers, and drug delivery vehicles. However, little is known about the characteristics of EVs secreted by hiPSCs and paracrine signaling during tissue morphogenesis and lineage specification. Methods: In this study, the physical and biological properties of EVs isolated from hiPSC-derived neural progenitors (ectoderm), hiPSC-derived cardiac cells (mesoderm), and the undifferentiated hiPSCs (healthy iPSK3 and Alzheimer&rsquo, s-associated SY-UBH lines) were analyzed. Results: Nanoparticle tracking analysis and electron microscopy results indicate that hiPSC-derived EVs have an average size of 100&ndash, 250 nm. Immunoblot analyses confirmed the enrichment of exosomal markers Alix, CD63, TSG101, and Hsc70 in the purified EV preparations. MicroRNAs including miR-133, miR-155, miR-221, and miR-34a were differently expressed in the EVs isolated from distinct hiPSC lineages. Treatment of cortical spheroids with hiPSC-EVs in vitro resulted in enhanced cell proliferation (indicated by BrdU+ cells) and axonal growth (indicated by &beta, tubulin III staining). Furthermore, hiPSC-derived EVs exhibited neural protective abilities in A&beta, 42 oligomer-treated cultures, enhancing cell viability and reducing oxidative stress. Our results demonstrate that the paracrine signaling provided by tissue context-dependent EVs derived from hiPSCs elicit distinct responses to impact the physiological state of cortical spheroids. Overall, this study advances our understanding of cell‒cell communication in the stem cell microenvironment and provides possible therapeutic options for treating neural degeneration.
- Published
- 2019
39. Extraction of Extracellular Vesicles from Whole Tissue
- Author
-
David G. Meckes, James Olcese, and Stephanie N. Hurwitz
- Subjects
0301 basic medicine ,General Chemical Engineering ,Cell ,Extracellular vesicles ,Article ,Mass Spectrometry ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,Extracellular Vesicles ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,General Immunology and Microbiology ,Chemistry ,Vesicle ,General Neuroscience ,Neurodegeneration ,Brain ,Proteins ,Extracellular vesicle ,medicine.disease ,Microvesicles ,Cell biology ,Microscopy, Electron ,030104 developmental biology ,medicine.anatomical_structure ,030217 neurology & neurosurgery ,Ex vivo - Abstract
Circulating and interstitial small membrane-bound extracellular vesicles (EVs) represent promising targets for the development of novel diagnostic or prognostic biomarker assays, and likely serve as important players in the progression of a vast spectrum of diseases. Current research is focused on the characterization of vesicles secreted from multiple cell and tissue types in order to better understand the role of EVs in the pathogenesis of conditions including neurodegeneration, inflammation, and cancer. However, globally consistent and reproducible techniques to isolate and purify vesicles remain in progress. Moreover, methods for extraction of EVs from solid tissue ex vivo are scarcely described. Here, we provide a detailed protocol for extracting small EVs of interest from whole fresh or frozen tissues, including brain and tumor specimens, for further characterization. We demonstrate the adaptability of this method for multiple downstream analyses, including electron microscopy and immunophenotypic characterization of vesicles, as well as quantitative mass spectrometry of EV proteins.
- Published
- 2019
40. Proteomic profiling of NCI-60 extracellular vesicles uncovers common protein cargo and cancer type-specific biomarkers
- Author
-
Mark A. Rider, Stephanie N. Hurwitz, Xia Liu, Rakesh K. Singh, David G. Meckes, and Joseph L. Bundy
- Subjects
Proteomics ,0301 basic medicine ,exosomes ,Biology ,Extracellular Vesicles ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Biomarkers, Tumor ,Humans ,Secretion ,Proteomic Profile ,Proteomic Profiling ,Vesicle ,biomarkers ,co-inertia ,Microvesicles ,3. Good health ,Cell biology ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Proteome ,Rab ,microvesicles ,Research Paper - Abstract
Packed with biological information, extracellular vesicles (EVs) offer exciting promise for biomarker discovery and applications in therapeutics and non-invasive diagnostics. Currently, our understanding of EV contents is confined by the limited cells from which vesicles have been characterized utilizing the same enrichment method. Using sixty cell lines from the National Cancer Institute (NCI-60), here we provide the largest proteomic profile of EVs in a single study, identifying 6,071 proteins with 213 common to all isolates. Proteins included established EV markers, and vesicular trafficking proteins such as Rab GTPases and tetraspanins. Differentially-expressed proteins offer potential for cancer diagnosis and prognosis. Network analysis of vesicle quantity and proteomes identified EV components associated with vesicle secretion, including CD81, CD63, syntenin-1, VAMP3, Rab GTPases, and integrins. Integration of vesicle proteomes with whole-cell molecular profiles revealed similarities, suggesting EVs provide a reliable reflection of their progenitor cell content, and are therefore excellent indicators of disease.
- Published
- 2016
41. Transmembrane Domains Mediate Intra- and Extracellular Trafficking of Epstein-Barr Virus Latent Membrane Protein 1
- Author
-
Deanna C. Tremblay, Xia Liu, Stephanie N. Hurwitz, Lauren A. Howell, Mujeeb R. Cheerathodi, David G. Meckes, and Dingani Nkosi
- Subjects
0301 basic medicine ,Herpesvirus 4, Human ,Viral protein ,DNA Mutational Analysis ,Immunology ,Biology ,Exosomes ,medicine.disease_cause ,Microbiology ,Exosome ,Viral Matrix Proteins ,03 medical and health sciences ,hemic and lymphatic diseases ,Virology ,otorhinolaryngologic diseases ,medicine ,Humans ,Secretion ,Endoplasmic reticulum ,Epstein–Barr virus latent membrane protein 1 ,Extracellular vesicle ,Microvesicles ,Virus-Cell Interactions ,Cell biology ,Protein Transport ,stomatognathic diseases ,Transmembrane domain ,HEK293 Cells ,030104 developmental biology ,Insect Science - Abstract
EBV latent membrane protein 1 (LMP1) is released from latently infected tumor cells in small membrane-enclosed extracellular vesicles (EVs). Accumulating evidence suggests that LMP1 is a major driver of EV content and functions. LMP1-modified EVs have been shown to influence recipient cell growth, migration, differentiation, and regulation of immune cell function. Despite the significance of LMP1-modified exosomes, very little is known about how this viral protein enters or manipulates the host EV pathway. In this study, LMP1 deletion mutants were generated to assess protein regions required for EV trafficking. Following transfection of LMP1 or mutant plasmids, EVs were collected by differential centrifugation, and the levels of specific cargo were evaluated by immunoblot analysis. The results demonstrate that, together, the N terminus and transmembrane region 1 of LMP1 are sufficient for efficient sorting into EVs. Consistent with these findings, a mutant lacking the N terminus and transmembrane domains 1 through 4 (TM5-6) failed to be packaged into EVs, and exhibited higher colocalization with endoplasmic reticulum and early endosome markers than the wild-type protein. Surprisingly, TM5-6 maintained the ability to colocalize and form a complex with CD63, an abundant exosome protein that is important for the incorporation of LMP1 into EVs. Other mutations within LMP1 resulted in enhanced levels of secretion, pointing to potential positive and negative regulatory mechanisms for extracellular vesicle sorting of LMP1. These data suggest new functions of the N terminus and transmembrane domains in LMP1 intra- and extracellular trafficking that are likely downstream of an interaction with CD63.IMPORTANCE EBV infection contributes to the development of cancers, such as nasopharyngeal carcinoma, Burkitt lymphoma, Hodgkin's disease, and posttransplant lymphomas, in immunocompromised or genetically susceptible individuals. LMP1 is an important viral protein expressed by EBV in these cancers. LMP1 is secreted in extracellular vesicles (EVs), and the transfer of LMP1-modified EVs to uninfected cells can alter their physiology. Understanding the cellular machinery responsible for sorting LMP1 into EVs is limited, despite the importance of LMP1-modified EVs. Here, we illustrate the roles of different regions of LMP1 in EV packaging. Our results show that the N terminus and TM1 are sufficient to drive LMP1 EV trafficking. We further show the existence of potential positive and negative regulatory mechanisms for LMP1 vesicle sorting. These findings provide a better basis for future investigations to identify the mechanisms of LMP1 targeting to EVs, which could have broad implications in understanding EV cargo sorting.
- Published
- 2018
42. Tetraspanin CD63 Bridges Autophagic and Endosomal Processes To Regulate Exosomal Secretion and Intracellular Signaling of Epstein-Barr Virus LMP1
- Author
-
Mujeeb R. Cheerathodi, Stephanie N. Hurwitz, Sara B. York, David G. Meckes, and Dingani Nkosi
- Subjects
0301 basic medicine ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cell signaling ,Tetraspanins ,Viral protein ,Immunology ,Endosomes ,Biology ,Exosomes ,medicine.disease_cause ,Microbiology ,Exocytosis ,Viral Matrix Proteins ,03 medical and health sciences ,Microscopy, Electron, Transmission ,Virology ,Autophagy ,medicine ,Humans ,Secretion ,Herpesviridae ,Exosomal secretion ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Sirolimus ,Tetraspanin 30 ,Secretory Vesicles ,TOR Serine-Threonine Kinases ,Virus Assembly ,Trehalose ,Extracellular vesicle ,Virus-Cell Interactions ,Cell biology ,Protein Transport ,HEK293 Cells ,030104 developmental biology ,Insect Science ,Vacuoles ,Intracellular ,Protein Binding ,Signal Transduction - Abstract
The tetraspanin protein CD63 has been recently described as a key factor in extracellular vesicle (EV) production and endosomal cargo sorting. In the context of Epstein-Barr virus (EBV) infection, CD63 is required for the efficient packaging of the major viral oncoprotein latent membrane protein 1 (LMP1) into exosomes and other EV populations and acts as a negative regulator of LMP1 intracellular signaling. Accumulating evidence has also pointed to intersections of the endosomal and autophagy pathways in maintaining cellular secretory processes and as sites for viral assembly and replication. Indeed, LMP1 can activate the mammalian target of rapamycin (mTOR) pathway to suppress host cell autophagy and facilitate cell growth and proliferation. Despite the growing recognition of cross talk between endosomes and autophagosomes and its relevance to viral infection, little is understood about the molecular mechanisms governing endosomal and autophagy convergence. Here, we demonstrate that CD63-dependent vesicle protein secretion directly opposes intracellular signaling activation downstream of LMP1, including mTOR-associated proteins. Conversely, disruption of normal autolysosomal processes increases LMP1 secretion and dampens signal transduction by the viral protein. Increases in mTOR activation following CD63 knockout are coincident with the development of serum-dependent autophagic vacuoles that are acidified in the presence of high LMP1 levels. Altogether, these findings suggest a key role of CD63 in regulating the interactions between endosomal and autophagy processes and limiting cellular signaling activity in both noninfected and virally infected cells. IMPORTANCE The close connection between extracellular vesicles and viruses is becoming rapidly and more widely appreciated. EBV, a human gamma herpesvirus that contributes to the progression of a multitude of lymphomas and carcinomas in immunocompromised or genetically susceptible populations, packages its major oncoprotein, LMP1, into vesicles for secretion. We have recently described a role of the host cell protein CD63 in regulating intracellular signaling of the viral oncoprotein by shuttling LMP1 into exosomes. Here, we provide strong evidence of the utility of CD63-dependent EVs in regulating global intracellular signaling, including mTOR activation by LMP1. We also demonstrate a key role of CD63 in coordinating endosomal and autophagic processes to regulate LMP1 levels within the cell. Overall, this study offers new insights into the complex intersection of cellular secretory and degradative mechanisms and the implications of these processes in viral replication.
- Published
- 2018
43. Extracellular Vesicle Biogenesis in Cancer
- Author
-
Stephanie N. Hurwitz and David G. Meckes
- Subjects
0301 basic medicine ,Cell ,Cancer ,Context (language use) ,Extracellular vesicle ,Biology ,medicine.disease ,Extracellular vesicles ,Cell biology ,Human tumor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Biogenesis - Abstract
Extracellular vesicles (EVs) represent a broad collection of membrane-enclosed sacs released from cells that contain biologically active cargo and cell type–specific and disease type–specific molecular information. Furthermore, EVs have been demonstrated to perpetuate the pathogenesis and progression of disease through manipulation of cells in the surrounding microenvironment. Based on the unique properties of EVs, there is tremendous interest in developing EV-based diagnostic and therapeutic platforms for cancer and many other diseases. However, before the full medical utility of EVs can be reached, a better understanding of mechanisms driving EV biogenesis must be obtained. Here, we will highlight the current understanding of EV biogenesis in the context of cancer, discuss recent advances in the field, and provide evidence for the utility of human tumor viruses in studying EV biogenesis and trafficking.
- Published
- 2018
44. List of Contributors
- Author
-
Maria Abreu, Aamir Ahmad, Rebaz Ahmed, Mansoor M. Amiji, Narsireddy Amreddy, Vincent Bernard, Sagar Bhayana, Cesar M. Castro, Zhenfeng Duan, Scott W. Ferguson, Cassandra Garbutt, Allison Gillaspy, Michael W. Graner, Mei He, Philip Hochendoner, Francis Hornicek, Stephanie N. Hurwitz, Hyungsoon Im, Naduparambil K. Jacob, Naureen Javeed, Ali S. Khan, Mohammad A. Khan, Salma Khan, Moh’d Khushman, Hakho Lee, Luize G. Lima, Jinjie Ling, Emil Lou, Anirban Maitra, David G. Meckes Jr., Jake S. Megna, Li Min, Andreas Möller, Katherine Moxley, Debabrata Mukhopadhyay, Anupama Munshi, Phillip B. Munson, Juliane Nguyen, Neha N. Parayath, Girijesh K. Patel, Mary C. Patton, Rajagopal Ramesh, Mohammed A. Razaq, Arti Shukla, Alfred A. Simental, Ajay P. Singh, Seema Singh, William Sperduto, Akhil Srivastava, Sanjeev K. Srivastava, Subbaya Subramanian, Mei-Ju Su, Yuki Takahashi, Yoshinobu Takakura, Rheal Towner, Malav S. Trivedi, Marshleen Yadav, Katherine S. Yang, Sara Yousuf, Liang Zhang, Yan D. Zhao, Zheng Zhao, and Haseeb Zubair
- Published
- 2018
45. Exosomal Communication Goes Viral
- Author
-
David G. Meckes
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cell signaling ,Viral matrix protein ,viruses ,Immunology ,Context (language use) ,Cell Communication ,Biology ,Exosomes ,Models, Biological ,Microbiology ,Exosome ,Virus ,Microvesicles ,Cell biology ,Viral Matrix Proteins ,Immune system ,Virology ,Insect Science ,Host-Pathogen Interactions ,Tumor Virus ,Humans ,Gems ,Immune Evasion - Abstract
Exosomes are small vesicles secreted from cells that participate in intercellular communication events. Accumulating evidence demonstrates that host exosome pathways are hijacked by viruses and that virally modified exosomes contribute to virus spread and immune evasion. In the case of tumor viruses, recent findings suggest that alterations in normal exosome biology may promote the development and progression of cancer. These studies will be discussed in the context of our current knowledge of Epstein-Barr virus (EBV)-modified exosomes.
- Published
- 2015
46. Erratum to: An Adaptable Polyethylene Glycol-Based Workflow for Proteomic Analysis of Extracellular Vesicles
- Author
-
Stephanie N, Hurwitz and David G, Meckes
- Published
- 2017
47. The interactome of EBV LMP1 evaluated by proximity-based BioID approach
- Author
-
Mujeeb R. Cheerathodi, Fanxiu Zhu, Stephanie N. Hurwitz, Dingani Nkosi, Xia Liu, Lauren A. Howell, David G. Meckes, Mark A. Rider, and Deanna C. Tremblay
- Subjects
0301 basic medicine ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Syntenins ,Sorting Nexins ,Biotin ,Cell Cycle Proteins ,Biology ,Proteomics ,medicine.disease_cause ,Exosomes ,Interactome ,Mass Spectrometry ,Article ,Protein–protein interaction ,Viral Matrix Proteins ,03 medical and health sciences ,Virology ,Protein targeting ,Protein Interaction Mapping ,medicine ,otorhinolaryngologic diseases ,TSG101 ,Humans ,Endosomal Sorting Complexes Required for Transport ,Tetraspanin 30 ,Calcium-Binding Proteins ,Microvesicles ,Cell biology ,stomatognathic diseases ,030104 developmental biology ,Host-Pathogen Interactions ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
Epstein-Barr virus LMP1 is an oncoprotein required for immortalizing B lymphocytes and also plays important roles in transforming non-lymphoid tissue. The discovery of LMP1 protein interactions will likely generate targets to treat EBV-associated cancers. Here, we define the broader LMP1 interactome using the recently developed BioID method. Combined with mass spectrometry, we identified over 1,000 proteins across seven independent experiments with direct or indirect relationships to LMP1. Pathway analysis suggests that a significant number of the proteins identified are involved in signal transduction and protein or vesicle trafficking. Interestingly, a large number of proteins thought to be important in the formation of exosomes and protein targeting were recognized as probable LMP1 interacting partners, including CD63, syntenin-1, ALIX, TSG101, HRS, CHMPs, and sorting nexins. Therefore, it is likely that LMP1 modifies protein trafficking and exosome biogenesis pathways. In support of this, knock-down of syntenin-1 and ALIX resulted in reduced exosomal LMP1.
- Published
- 2017
48. An Adaptable Polyethylene Glycol-Based Workflow for Proteomic Analysis of Extracellular Vesicles
- Author
-
Stephanie N, Hurwitz and David G, Meckes
- Subjects
Proteomics ,Extracellular Vesicles ,Proteome ,Blotting, Western ,Exosomes ,Ultracentrifugation ,Polyethylene Glycols - Abstract
Extracellular vesicles (EVs), including exosomes are endocytically derived nanovesicles expelled from cells that contain molecular information in the form of lipids, proteins, and nucleic acids. Transfer of this information to other cells in local or distant microenvironments facilitates cell-to-cell communication. Importantly, diseased cells release exosomes containing specific cargo that may contribute to pathology and can be harnessed for diagnostic or prognostic use. The broad potential medical utility of exosomes has fueled rapidly expanding research on understanding the composition and functions of exosomes in normal and pathological conditions. Here, we provide a complete workflow for purifying exosome-sized vesicles from biological fluids for in-depth proteomic analyses. Moreover, this polyethylene glycol-based method is efficient, highly adaptable, and compatible with a variety of downstream applications.
- Published
- 2017
49. CD63 Regulates Epstein-Barr Virus LMP1 Exosomal Packaging, Enhancement of Vesicle Production, and Noncanonical NF-κB Signaling
- Author
-
David G. Meckes, Meghan M. Conlon, Stephanie N. Hurwitz, Sara B. York, Dingani Nkosi, Deanna C. Tremblay, and Xia Liu
- Subjects
0301 basic medicine ,Cell signaling ,Herpesvirus 4, Human ,Endosome ,Immunology ,Biology ,Exosomes ,Microbiology ,Transformation and Oncogenesis ,Viral Matrix Proteins ,03 medical and health sciences ,Membrane Microdomains ,Virology ,otorhinolaryngologic diseases ,Animals ,Humans ,Secretion ,Lipid raft ,Tumor microenvironment ,Tetraspanin 30 ,Secretory Vesicles ,NF-kappa B ,Extracellular vesicle ,Microvesicles ,Cell biology ,Rats ,stomatognathic diseases ,Protein Transport ,030104 developmental biology ,HEK293 Cells ,Insect Science ,Cancer research ,Signal transduction ,Signal Transduction - Abstract
Latent membrane protein 1 (LMP1) is an Epstein-Barr virus (EBV)-encoded oncoprotein that is packaged into small extracellular vesicles (EVs) called exosomes. Trafficking of LMP1 into multivesicular bodies (MVBs) alters the content and function of exosomes. LMP1-modified exosomes enhance the growth, migration, and invasion of malignant cells, demonstrating the capacity to manipulate the tumor microenvironment and enhance the progression of EBV-associated cancers. Despite the growing evidence surrounding the significance of LMP1-modified exosomes in cancer, very little is understood about the mechanisms that orchestrate LMP1 incorporation into these vesicles. Recently, LMP1 was shown to be copurified with CD63, a conserved tetraspanin protein enriched in late endosomal and lysosomal compartments. Here, we demonstrate the importance of CD63 presence for exosomal packaging of LMP1. Nanoparticle tracking analysis and gradient purification revealed an increase in extracellular vesicle secretion and exosomal proteins following LMP1 expression. Immunoisolation of CD63-positive exosomes exhibited accumulation of LMP1 in this vesicle population. Functionally, CRISPR/Cas9 knockout of CD63 resulted in a reduction of LMP1-induced particle secretion. Furthermore, LMP1 packaging was severely impaired in CD63 knockout cells, concomitant with a disruption in the perinuclear localization of LMP1. Importantly, LMP1 trafficking to lipid rafts and activation of NF-κB and PI3K/Akt pathways remained intact following CD63 knockout, while mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) and noncanonical NF-κB activation were observed to be increased. These results suggest that CD63 is a critical player in LMP1 exosomal trafficking and LMP1-mediated enhancement of exosome production and may play further roles in limiting downstream LMP1 signaling. IMPORTANCE EBV is a ubiquitous gamma herpesvirus linked to malignancies such as nasopharyngeal carcinoma, Burkitt's lymphoma, and Hodgkin's lymphoma. In the context of cancer, EBV hijacks the exosomal pathway to modulate cell-to-cell signaling by secreting viral components such as an oncoprotein, LMP1, into host cell membrane-bound EVs. Trafficking of LMP1 into exosomes is associated with increased oncogenicity of these secreted vesicles. However, we have only a limited understanding of the mechanisms surrounding exosomal cargo packaging, including viral proteins. Here, we describe a role of LMP1 in EV production that requires CD63 and provide an extensive demonstration of CD63-mediated exosomal LMP1 release that is distinct from lipid raft trafficking. Finally, we present further evidence of the role of CD63 in limiting LMP1-induced noncanonical NF-κB and ERK activation. Our findings have implications for future investigations of physiological and pathological mechanisms of exosome biogenesis, protein trafficking, and signal transduction, especially in viral-associated tumorigenesis.
- Published
- 2017
50. Erratum to: An Adaptable Polyethylene Glycol-Based Workflow for Proteomic Analysis of Extracellular Vesicles
- Author
-
David G. Meckes and Stephanie N. Hurwitz
- Subjects
chemistry.chemical_compound ,Workflow ,Materials science ,chemistry ,Nanotechnology ,Polyethylene glycol ,Extracellular vesicles - Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.