Spelling suggestions: "subject:"exosomes"" "subject:"hexosomes""
121 |
HCV-associated Exosomes Promote Myeloid-Derived Suppressor Cell Expansion via Inhibiting miR-124 to Regulate T Follicular Cell Differentiation and FunctionWang, Ling, Cao, Dechao, Wang, Ling, Zhao, Juan, Nguyen, Lam Nhat, Dang, Xindi, Ji, Yingjie, Wu, Xiao Y., Morrison, Zheng D., Xie, Qian, El Gazzar, Mohamed, Ning, Shunbin, Moorman, Jonathon P., Yao, Zhi Q. 11 September 2018 (has links) (PDF)
Virus-infected cells can regulate non-permissive bystander cells, but the precise mechanisms remain incompletely understood. Here we report that this process can be mediated by transfer of viral RNA-loaded exosomes shed from infected cells to myeloid-derived suppressor cells (MDSCs), which in turn regulate the differentiation and function of T cells during viral infection. Specifically, we demonstrated that patients with chronic hepatitis C virus (HCV) infection exhibited significant increases in T follicular regulatory (TFR) cells and decreases in T follicular helper (TFH) cells. These MDSC-mediated T-cell dysregulations resulted in an increased ratio of TFR/TFH and IL-10 production in peripheral blood. Specifically, co-culture of MDSCs derived from HCV patients with healthy peripheral blood mononuclear cells (PBMCs) induced expansion of TFR, whereas depletion of MDSCs from PBMCs of HCV patients reduced the increases in TFR frequency and IL-10 production, and promoted the differentiation of IFN-γ-producing TFH cells. Importantly, we found that exosomes isolated from the plasma of HCV patients and supernatant of HCV-infected hepatocytes could drive monocytic myeloid cell differentiation into MDSCs. These exosomes were enriched in tetraspanins, such as CD63 and CD81, and contained HCV RNA, but exosomes isolated from patients with antiviral treatment contained no HCV RNA and could not induce MDSC differentiation. Notably, these HCV RNA-containing exosomes (HCV-Exo) were sufficient to induce MDSCs. Furthermore, incubation of healthy myeloid cells with these HCV-Exo inhibited the expression of miR−124, whereas reconstitution of PBMCs with miR−124 abolished the effects of HCV−Exo on MDSC induction. Taken together, these results indicate that HCV-associated exosomes can transfer immunomodulatory viral RNA from infected cells to neighboring immune cells and trigger MDSC expansion, which subsequently promotes TFR differentiation and inhibits TFH function. This study reveals a previously unrecognized path that represents a novel mechanism of immune dysregulation during chronic viral infection.
|
122 |
INVESTIGATION OF NOVEL THERAPIES AND DELIVERY SYSTEMS FOR TREATMENT OF HEPATOCELLULAR CARCINOMABadawi, Mohamed A. January 2017 (has links)
No description available.
|
123 |
Targeted Delivery of a Therapeutic Protein for the Treatment of Alzheimer's DiseaseHolman, Heather 01 January 2018 (has links) (PDF)
Neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease are linked to mitochondrial dysfunction and the underexpression of TOM40, a protein with chaperone-like qualities that is responsible for transporting precursor proteins into the mitochondria. Overexpression of TOM40 is reported to partially restore mitochondrial dysfunction and decrease the accumulation of neurotoxic aggregates of α-synuclein. Our goal is to develop an effective method for delivery of TOM40 protein to the brain.
Previous studies have used lentiviruses to carry TOM40 into the hippocampus of α-synuclein transgenic mice. The disadvantage of lentiviral transfection is the random insertions of the target gene into the host genome, which could cause toxic effects. Synthetic phospholipid vesicles containing TOM40 were considered as an alternative delivery method, but these "liposomes" elicit not only toxicity, but also an immune response. Thus, development of a safer delivery method of TOM40 protein is needed. We investigated exosomes, which are extracellular vesicles originating from multivesicular endosomes filled with protein, lipid, or RNA cargoes for cell-cell communication. Since exosomes are created from host cells, they are non-immunogenic and may be a more desirable method.
Expression constructs have been made for the production of TOM40 protein within or on the surface of exosomes. In order to target the delivery of TOM40 to the brain, we attached peptides to the surface of the exosomes, which specifically interact with receptors on neural cells. We attempted to confirm the functionality of the expression constructs through immunocytochemistry followed by flow cytometry and Western blotting.
|
124 |
Utilisation des vésicules extracellulaires de sérum comme véhicule de livraison du système CRISPR-Cas9 pour traiter la Dystrophie Musculaire de DuchenneFortin-Archambault, Annabelle 18 October 2022 (has links)
La dystrophie musculaire de Duchenne est une maladie génétique qui résulte de diverses mutations dans le gène DMD, codant pour la protéine dystrophine. 70% des patients ont une délétion d'exons ou de parties d'exons provoquant un changement dans le cadre de lecture, résultant en l'apparition d'un codon stop et en l'absence de la protéine dystrophine. Plusieurs traitements potentiels ont été explorés dans les dernières années pour cette maladie, dont le système CRISPR-Cas9, un outil génétique permettant d'éliminer un segment d'ADN à l'aide de la protéine nucléase Cas9 et de deux guides d'ARN ciblant des séquences précises d'ADN. Le plus grand défi avec l'utilisation de cette technologie est sa livraison in vivo. Les vésicules extracellulaires sont des particules membranaires lipidiques qui jouent un rôle dans la communication intercellulaire et sont retrouvées dans tous les biofluides chez les mammifères. Elles pourraient donc être une alternative intéressante pour la livraison du système CRISPR-Cas9. J'ai participé à des travaux de purification de vésicules extracellulaires de sérum par chromatographie par exclusion de taille. Ces vésicules extracellulaires ont été chargées avec la protéine Cas9 et des guides ARN, puis, des injections intramusculaires ont été effectuées dans le Tibialis anterior de trois lignées de souris (Ai9, RAG-mdx et mdx/hDMD) pour établir l'efficacité du traitement. Les résultats ont montré que les vésicules extracellulaires chargées avec la Cas9 et des guides d'ARN provoquent une édition de l'ADN efficace dans le Tibialis anterior des trois lignées de souris utilisées et la restauration de l'expression de la protéine dystrophine dans les fibres musculaires du Tibialis anterior des souris RAG-mdx, modèle pour la dystrophie musculaire de Duchenne. Le traitement a ensuite été modifié pour permettre le ciblage des vésicules extracellulaires aux organes affectés par la dystrophie musculaire de Duchenne, soit le cœur et les muscles squelettiques. Des peptides de ciblage ont été sélectionnés dans la littérature et insérés dans la membrane des vésicules extracellulaires marquées de façon fluorescente à l'aide d'un segment lipidique stéaryl. Les résultats de l'expérience effectuée avec les vésicules extracellulaires ciblées n'ont pas été concluants en raison d'un marquage mal adapté des vésicules injectées, mais de futures expériences permettront d'élucider leur efficacité. L'ajustement du traitement pour permettre une injection systémique rejoignant le cœur et les muscles squelettiques est indispensable à l'application de celui-ci à la clinique. / Duchenne muscular dystrophy is a genetic disease that affects one in 3500 boys and results from mutations in the DMD gene, which codes for dystrophin protein. 70% of patients have an exon deletion, which results in a shift in the reading frame, the apparition of a stop codon, and the absence of the dystrophin protein. Many different potential treatments have been explored for Duchenne muscular dystrophy, including the CRISPRCas9 system. This technology allows for the modification of genomic DNA through a Cas9 nuclease and two guide RNAs designed to target a specific DNA sequence. The biggest challenge with using the CRISPR system is delivery. The classic vectors for CRISPR, such as AAV, can cause many adverse effects like immunological responses. Extracellular vesicles are membranous particles that play a role in intercellular communication and are found in all mammalian biofluids. They are thus an interesting alternative for the delivery of the Cas9 protein and its guide RNAs. I have participated in a research project aiming to purify serum extracellular vesicles by size-exclusion chromatography and to load them with Cas9 and two guide RNAs. These extracellular vesicles were then injected intramuscularly into the Tibialis anterior muscles of three mouse strains (Ai9, RAG-mdx and mdx/hDMD) to assess treatment efficiency. The injection of Cas9 and guide RNA-loaded extracellular vesicles produced efficient gene editing as well as dystrophin expression restauration. To modify the treatment for systemic injection, targeting peptides were added to EV membrane through a lipid stearyl segment. This was done to target the extracellular vesicles to Duchenne muscular dystrophy-affected organs: heart and skeletal muscles. Results of the targeted-extracellular vesicle experiment were inconclusive, however, with more experiments, the efficacy of the targeting peptides should be determinable. It is essential to adjust this treatment to allow for targeted systemic delivery for it to be applicable to the clinic.
|
125 |
The therapeutic/anti-carcinogenic effect of cord blood stem cells-derived exosomes in malignant melanomaNaeem, Parisa January 2022 (has links)
Malignant melanoma is an invasive type of skin cancer with high mortality rates, if not detected promptly. The mortality trends are generally linked to multiple dysplastic nevi, positive family history, genetic susceptibility and phenotypic features including fair skin, freckles, numerous atypical nevi, light coloured hair and eyes, inability to tan and prolonged exposure to ultraviolet radiation B (UVB). To date, the major anti-cancer therapeutics for melanoma include surgery, chemotherapy, radiotherapy, and immunotherapy. Recently, extracellular vesicles, especially exosomes, have been highlighted for their therapeutic benefits in numerous chronic diseases such as cancer. Exosomes display multifunctional properties, including inhibition of cancer cell proliferation and initiation of apoptosis. Hence, this study aimed to evaluate the genotoxicity and cytotoxicity of cord blood stem cell-derived (CBSC) exosomes on 6 samples of peripheral blood lymphocytes taken from healthy individuals and melanoma patients and on 3 samples of melanoma (CHL-1) cells. The limited number of samples was due to the time limitations and restrictions that were in place due to the COVID-19 pandemic. In this in vitro study, the optimal concentration of CBSC-derived exosomes (0, 100, 200, 300, 400 μg/ml protein at 24, 48 and 72h treatments) was confirmed by the CCK-8 assay.
CBSC exosomes (300 μg/ml) were used to treat lymphocytes and CHL-1 cells in the Comet assay and evaluated using the real-time polymerase chain reaction (qPCR) and Western blotting (WB). The data of the CCK-8 and Comet assays illustrated that exosomes exerted genotoxic effects on CHL-1 cells (CCK-8 assay, ****p < 0.0001), (Comet assay, *p <0.05, **p < 0.01). However, the data portraying a reduction in the viability of lymphocytes needs further investigation as the number of samples was limited, therefore, further clarification is required. Importantly, no significant adverse effect was observed in healthy lymphocytes when treated with the same exosomes (p = ns). When further challenged with UVA+B radiation, the exosomes did not induce any genoprotective effect on ROS-induced CHL-1 cells, compared to the positive control (p = ns). Our data insinuates that the damage might be caused by inducing apoptosis. The anti-tumourigenic potential of exosomes was observed by activating the p53-mediated apoptotic pathway in CHL-1 cells, up-regulating p53, p21 and caspase 3 and down-regulating BCL-2 at mRNA (**p < 0.01, ***p <0.001, ****p <0.0001) and protein levels (*p < 0.05, **p <0.01). The potency of CBSC exosomes in inhibiting cancer progression in CHL-1 cells whilst causing no harm to the healthy lymphocytes makes it an ideal potential candidate for anti-cancer therapy. More samples are required to evaluate the therapeutic effect of exosomes on lymphocytes from cancer patients to fully understand their mechanism of action.
|
126 |
Characterization of Histone H1 and Extracellular Vesicles by Mass SpectrometryHarshman, Sean William January 2013 (has links)
No description available.
|
127 |
Sex, pregnancy, and a great pair of genes: critical mediators in the development and prograssion of CNS autoimmune injuryGatson, NaTosha Na Chole 10 December 2007 (has links)
No description available.
|
128 |
Regulation of the tumour suppressor PTEN through exosomesGabriel, Kathleen 10 1900 (has links)
<p>PTEN is a potent tumour suppressor protein. Aggressive and metastatic prostate cancer (PC) is associated with a reduction or loss of PTEN expression. PTEN reduction often occurs without gene mutations, and its downregulation is not fully understood. Herein, we show that PTEN is incorporated in the cargo of exosomes derived from cancer cells, and this is an exclusive characteristic of cancer cells; normal cells do not incorporate PTEN in their exosomes. We found that this process is affected by the expression of oncogenes, with activation of oncogenic molecules leading to increased PTEN incorporation into exosomes. PTEN expressed in exosomes can be transferred to other cells that have a reduction or loss of PTEN expression. The transferred PTEN is active, as cells showed a substantial increase in phosphatase activity upon treatment with PTEN-bearing exosomes. PTEN transferred through exosomes is also competent to confer tumour-suppression activity to acceptor cells. After incubation with PTEN-bearing exosomes, recipient cells exhibited decreased AKT phosphorylation, changes in the expression of cell cycle mediators indicating cell cycle arrest, and decreased proliferation. These data suggest that exosomal PTEN may be able to compensate for PTEN loss in cancer cells, by transferring the active protein to cancer cells where it can then perform its role as a tumour suppressor.</p> / Master of Science (MSc)
|
129 |
Exploring Interactions Between Malignant Brain Cancer Cells and the Tumor Microenvironment Following High-Frequency Irreversible ElectroporationMurphy, Kelsey Rose 30 July 2024 (has links)
High-frequency irreversible electroporation (H-FIRE) is a novel tumor ablation therapeutic that applies bipolar, high-frequency pulsed electric fields to tumors, triggering the formation of irreversible membrane pores and to induce tumor cell death. H-FIRE has demonstrated pre-clinical and clinical utility as a therapeutic for brain tumors, including gliomas. H-FIRE has been shown to induce precise, uniform ablation within the tumor tissue, as well as local changes to the tumor microenvironment and systemic changes to the immune landscape. Namely, disruption of the peritumoral blood-brain barrier (BBB) following H-FIRE ablation of brain tumors, and infiltration and activation of the innate immune system are clinically observed following H-FIRE tumor ablation. Such effects persist long after death of the treated tumor, and therefore an understanding of the mechanisms underlying these local and systemic changes are critical for the development of H-FIRE. Using in vitro models of glioma and lung carcinoma-derived brain metastases, we investigate the interactions between cancer cells that have been ablated with H-FIRE and the brain tumor microenvironments. Specifically, we demonstrate that H-FIRE-treated cancer cells can recover treatment-induced damage and proliferative capacity after treatment with specific electric field doses, while higher doses inhibit such recovery. This suggests that after H-FIRE ablation of brain tumors, tumor cells can still secrete factors to trigger alterations in their local and systemic environments. We then specifically investigate the role of tumor-derived extracellular vesicles (TDEVs) in mediating these changes, namely pBBB disruption and changes in innate immunity. We find that, following H-FIRE ablation of brain cancer cells, treated cells immediately release TDEVs that disrupt the blood-brain barrier (BBB) endothelium in vitro, and are uniquely internalized by cerebral endothelial cells in vitro, despite reduced release of TDEVs after H-FIRE. We further demonstrate that H-FIRE significantly alters the proteomic payloads of TDEVs. When TDEVs released by sham- and H-FIRE-treated glioma cells are delivered to healthy rats, only TDEVs released by H-FIRE-ablated cells are retained in the brain, suggesting changes to TDEV organotropism after H-FIRE ablation of glioma. Further, once retained in the brain, these post-H-FIRE TDEVs cluster near cerebral endothelial cells, similarly to in vitro. Although the TDEVs released by H-FIRE ablated glioma cells do not disrupt the BBB in vivo, Iba1+ cells were increased in the brains of rats that received TDEVs released by H-FIRE-ablated glioma cells. Together, these data suggest that H-FIRE immediately alters the secretion and proteome of TDEVs, facilitating changes in TDEV organotropism and cellular tropism and immune cell recruitment to the tumor microenvironment. Together, this research indicates mechanisms by which tumor cells continue to modulate their local and systemic environments via the action of TDEVs, which is critical information for the continued development of H-FIRE and its optimization with adjuvant therapeutics for the treatment of malignant brain tumors. / Doctor of Philosophy / All cells secrete extracellular vesicles, which are packets of information that function as communication highways between cells. In cancer, tumor-derived extracellular vesicles (TDEVs) reprogram local and distant cells to support tumor growth. However, they have also been shown to change local and systemic functions, such as blood vessel function and immune response, after tumors are treated with therapeutics. Therefore, a full understanding of the role of TDEVs in how tumors communicate with the body after cancer treatment is necessary when developing new anti-cancer therapeutics. Here, in developing high-frequency irreversible electroporation (H-FIRE), a novel anti-tumor therapeutic for the treatment of malignant brain tumors, we explore how TDEVs released by brain cancer cells treated with H-FIRE interact with various cell types and structures in the body, and how these interactions may affect the response to treatment. Using a glioma model of primary brain cancer, and a lung carcinoma model of brain metastases, we first explore how tumor cells may be able to recover from damage after treatment with H-FIRE. We discover that brain cancer cells treated with specific doses of H-FIRE recover cell damage and continue to proliferate, but cells treated with higher doses of H-FIRE cannot recover these functions. The fact that tumor cells may be able to recover after H-FIRE suggests that cancer cells may still secrete factors, such as TDEVs, that interact with cells in the microenvironment after tumor treatment. We investigated the role of TDEVs released by brain cancer cells treated with H-FIRE to determine whether they cause changes in surrounding cells and structures in the brain cancer microenvironment. We determined that brain cancer cells treated with H-FIRE release TDEVs that carry proteins different from those carried by TDEVs routinely released by untreated cells. We further found that these TDEVs disrupt the blood-brain barrier (BBB) endothelium in vitro, and are uniquely internalized by cells of the endothelium. When these TDEVs were administered to the brains of healthy rats, they were retained in the brain, clustered near the endothelium, and recruited immune cells from circulation into the brain. Conversely, TDEVs that were routinely released from the brain cancer cells, in the absence of H-FIRE treatment, exhibited none of these functions. Taken together, these results show that H-FIRE changes TDEVs in numerous ways: after H-FIRE, the TDEVs may gravitate toward particular organs and cell types, and recruit immune cells. All of these changes can impact the overall therapeutic response after H-FIRE, and may also be specifically optimized and targeted with additional therapeutics to make H-FIRE more effective for brain cancer.
|
130 |
Utilisation des vésicules extracellulaires de sérum comme véhicule de livraison du système CRISPR-Cas9 pour traiter la Dystrophie Musculaire de DuchenneFortin-Archambault, Annabelle 14 November 2023 (has links)
La dystrophie musculaire de Duchenne est une maladie génétique qui résulte de diverses mutations dans le gène DMD, codant pour la protéine dystrophine. 70% des patients ont une délétion d'exons ou de parties d'exons provoquant un changement dans le cadre de lecture, résultant en l'apparition d'un codon stop et en l'absence de la protéine dystrophine. Plusieurs traitements potentiels ont été explorés dans les dernières années pour cette maladie, dont le système CRISPR-Cas9, un outil génétique permettant d'éliminer un segment d'ADN à l'aide de la protéine nucléase Cas9 et de deux guides d'ARN ciblant des séquences précises d'ADN. Le plus grand défi avec l'utilisation de cette technologie est sa livraison in vivo. Les vésicules extracellulaires sont des particules membranaires lipidiques qui jouent un rôle dans la communication intercellulaire et sont retrouvées dans tous les biofluides chez les mammifères. Elles pourraient donc être une alternative intéressante pour la livraison du système CRISPR-Cas9. J'ai participé à des travaux de purification de vésicules extracellulaires de sérum par chromatographie par exclusion de taille. Ces vésicules extracellulaires ont été chargées avec la protéine Cas9 et des guides ARN, puis, des injections intramusculaires ont été effectuées dans le Tibialis anterior de trois lignées de souris (Ai9, RAG-mdx et mdx/hDMD) pour établir l'efficacité du traitement. Les résultats ont montré que les vésicules extracellulaires chargées avec la Cas9 et des guides d'ARN provoquent une édition de l'ADN efficace dans le Tibialis anterior des trois lignées de souris utilisées et la restauration de l'expression de la protéine dystrophine dans les fibres musculaires du Tibialis anterior des souris RAG-mdx, modèle pour la dystrophie musculaire de Duchenne. Le traitement a ensuite été modifié pour permettre le ciblage des vésicules extracellulaires aux organes affectés par la dystrophie musculaire de Duchenne, soit le cœur et les muscles squelettiques. Des peptides de ciblage ont été sélectionnés dans la littérature et insérés dans la membrane des vésicules extracellulaires marquées de façon fluorescente à l'aide d'un segment lipidique stéaryl. Les résultats de l'expérience effectuée avec les vésicules extracellulaires ciblées n'ont pas été concluants en raison d'un marquage mal adapté des vésicules injectées, mais de futures expériences permettront d'élucider leur efficacité. L'ajustement du traitement pour permettre une injection systémique rejoignant le cœur et les muscles squelettiques est indispensable à l'application de celui-ci à la clinique. / Duchenne muscular dystrophy is a genetic disease that affects one in 3500 boys and results from mutations in the DMD gene, which codes for dystrophin protein. 70% of patients have an exon deletion, which results in a shift in the reading frame, the apparition of a stop codon, and the absence of the dystrophin protein. Many different potential treatments have been explored for Duchenne muscular dystrophy, including the CRISPRCas9 system. This technology allows for the modification of genomic DNA through a Cas9 nuclease and two guide RNAs designed to target a specific DNA sequence. The biggest challenge with using the CRISPR system is delivery. The classic vectors for CRISPR, such as AAV, can cause many adverse effects like immunological responses. Extracellular vesicles are membranous particles that play a role in intercellular communication and are found in all mammalian biofluids. They are thus an interesting alternative for the delivery of the Cas9 protein and its guide RNAs. I have participated in a research project aiming to purify serum extracellular vesicles by size-exclusion chromatography and to load them with Cas9 and two guide RNAs. These extracellular vesicles were then injected intramuscularly into the Tibialis anterior muscles of three mouse strains (Ai9, RAG-mdx and mdx/hDMD) to assess treatment efficiency. The injection of Cas9 and guide RNA-loaded extracellular vesicles produced efficient gene editing as well as dystrophin expression restauration. To modify the treatment for systemic injection, targeting peptides were added to EV membrane through a lipid stearyl segment. This was done to target the extracellular vesicles to Duchenne muscular dystrophy-affected organs: heart and skeletal muscles. Results of the targeted-extracellular vesicle experiment were inconclusive, however, with more experiments, the efficacy of the targeting peptides should be determinable. It is essential to adjust this treatment to allow for targeted systemic delivery for it to be applicable to the clinic.
|
Page generated in 0.0325 seconds