scispace - formally typeset
Search or ask a question

What is the frontier of knowledge in the use of exosomes in glioblastoma? 


Best insight from top research papers

The frontier of knowledge in the use of exosomes in glioblastoma lies in their role as potent carriers for targeted therapy and diagnostic markers. Exosomes, being small extracellular vesicles, facilitate intercellular communication and play crucial roles in tumor progression, immune tolerance, and drug resistance in gliomas. They are explored for their ability to deliver drugs effectively across the blood-brain barrier, a significant challenge in glioblastoma treatment. Moreover, exosomes are rich in biomolecules like proteins, nucleic acids, and microRNAs, making them valuable sources of diagnostic and prognostic markers for glioblastoma. The ongoing research focuses on harnessing exosomes' inherent properties for developing precise and biocompatible therapeutic strategies, marking a promising avenue in the emerging era of precision medicine.

Answers from top 5 papers

More filters
Papers (5)Insight
Exosomes, containing CD44, VEGFA, TGFβ1, THBS1, and SERPINE1, serve as early non-invasive predictors of disease state in differentiating high-grade from low-grade glioblastomas, particularly in blood serum samples.
Exosomes are explored as potential biomarkers for diagnosis, disease progression, and as delivery platforms for glioblastoma treatment, showcasing a frontier in precision medicine and targeted therapy.
Exosomes serve as promising non-invasive drug delivery tools to overcome the blood-brain barrier and target glioblastoma cells effectively, representing a frontier in glioblastoma therapy.
Exosomes in gliomas play a crucial role in tumor progression, immune evasion, and drug resistance. They offer potential as diagnostic biomarkers and drug delivery systems, advancing glioblastoma research and therapy.
Exosomes serve as promising drug carriers for treating glioblastoma due to their cell-targeting properties, stability, and potential diagnostic markers, advancing targeted therapy in GBM treatment.

Related Questions

Can exosomes be used for disc degeneration therapy?5 answersExosomes have shown potential for disc degeneration therapy. They can be released by cells and play a role in intercellular material transport and information transmission. Exosomes contain microRNAs that can regulate the physiological activities and functions of receptor cells. Studies have focused on the role of exosomes and exosomal microRNAs in the treatment of intervertebral disc degeneration (IVDD). Mesenchymal stem cell-derived exosomes have been shown to promote cell proliferation, tissue regeneration, and modulation of the inflammatory response in IVDD. Exosomes can be used as carriers to deliver their contents to target cells and regulate target cell activity. They have a low immunogenicity and no immune rejection, making them a promising therapeutic modality in IVDD. Exosomes can be integrated within a biomaterial delivery system to facilitate sustained delivery. Overall, exosomes hold promise for disc degeneration therapy by promoting regeneration and modulating the inflammatory response.
What is exosome?5 answersExosomes are nanosized membrane-bound vesicles that participate in intercellular communication and act as messengers in various pathological complications. They contain proteins, lipids, metabolites, miRNA, and DNA, and can transfer their contents to recipient cells. Exosomes are released from cells of different origins and can be found in various body fluids. They play a role in cancer progression, including uncontrolled cell growth, immune cell reprogramming, metastasis, and therapeutic resistance. Exosomes also have an impact on epigenetic alterations and can be used for cancer examination and research. Exosomes are involved in the life cycle of viruses such as HIV-1, hepatitis B and C, and SARS-CoV-2. They can transfer viral components to uninfected cells and have multidirectional effects on viral infections. Exosomes have potential applications in noninvasive diagnostics as biomarkers and as therapeutic agents, including genetically modified exosomes for antiviral vaccines. Exosomes have therapeutic potential for the treatment of various pathologies, including autoimmune, cardiovascular, ocular, and nervous disorders. They play a role in cell signaling and intercellular communication, and can penetrate the blood-brain barrier and maintain homeostasis in the central nervous system. Exosomes can be used as biomarkers for disease monitoring, early diagnosis, and treatment optimization. Exosomes are a subset of extracellular vesicles that transport functional molecules and have been shown to be effective diagnostic, prognostic, and predictive biomarkers in cancer. They have generated interest in cancer treatment as biological carriers for drugs, inhibitors, antibodies, and microRNA. Exosomes can serve as valuable biomarkers due to their specific content originating from their cells of origin.
How are exosomes used in cancer treatment?4 answersExosomes are being used in cancer treatment in various ways. They have been found to play a role in tumor progression, metastasis, and immune suppression. Exosomes can act as biomarkers for cancer diagnosis, prognosis, and prediction. They can also be used as drug delivery vehicles for cancer therapy. Engineered exosomes have advantages such as low immunogenicity, natural targeting ability, and suitable mechanical properties, making them reliable and safe drug carriers. Exosomes can be loaded with drugs, inhibitors, antibodies, and microRNA to target cancer cells. Additionally, exosomes derived from cancer stem cells have been found to play a significant role in tumor development, progression, and immune evasion. Understanding the characteristics and functions of exosomes may lead to the development of new diagnostic tools and therapies to prevent tumor resistance and relapse.
What is the efficacy of exosomes?3 answersExosomes have shown efficacy in various therapeutic applications, including musculoskeletal regeneration, wound healing, female infertility, cardiac recovery, immunomodulation, neurological disease, and metabolic regulation. They have been explored as delivery vehicles for chemotherapeutic drugs, such as 5-Fluorouracil, and have demonstrated enhanced drug release and cytotoxic effects in cancer cells. Exosomes derived from immune cells have potential applications in immunotherapy and immune regulation, including autoimmune connective tissue diseases. In the male reproductive system, exosomes produced by sertoli cells play a crucial role in supporting germ cell development and have potential diagnostic and regenerative applications in male infertility. Additionally, stem cell-derived exosomes have shown promise in wound healing by promoting angiogenesis, cell proliferation, and balancing inflammatory responses. Overall, exosomes have demonstrated efficacy in various therapeutic areas and hold potential for future clinical applications.
What is the role of exosomes in cancer?5 answersExosomes play a significant role in cancer by facilitating intercellular communication and influencing tumor growth and progression. They are small membrane-bound vesicles released by various cells, including cancer cells, and contain bioactive molecules such as proteins, lipids, and nucleic acids. Exosomes can affect the cells in the tumor microenvironment, including immune cells and pancreatic stellate cells, promoting tumor growth and the production of extracellular matrix components. Additionally, exosomes can be used as potential diagnostic tools for cancer, as their presence and molecular signature in body fluids can aid in early-stage diagnosis and monitoring. Furthermore, exosomes have been explored as a therapeutic tool for cancer treatment, including drug delivery systems, with the potential to enhance the effectiveness of chemotherapy drugs. Overall, exosomes contribute to the development, progression, diagnosis, monitoring, and treatment of cancer.
How can exosomes be used for diagnosis and treatment of dementia?5 answersExosomes can be used for the diagnosis and treatment of dementia by serving as diagnostic markers and therapeutic agents. Exosomes are involved in the pathogenesis of various subtypes of dementia, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), vascular dementia (VaD), dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD). They transport toxic proteins such as amyloid beta (Aβ), tau, and α-synuclein, which are characteristic of these diseases. Exosomal microRNAs (miR) and proteins reflect the disease state and can be used as diagnostic markers for different types and stages of dementia. Additionally, exosomes derived from mesenchymal stem cells (MSCs) have shown potential in ameliorating disease pathology and improving cognitive function in AD, PD, and VAD. Furthermore, exosomes can be loaded with active pharmaceutical ingredients (APIs) to specifically target and treat dementia. The detection and analysis of exosomes in biological fluids offer a multicomponent diagnostic readout for dementia. Overall, exosomes have the potential to revolutionize the diagnosis and treatment of dementia.

See what other people are reading

What are the links between allergies and neuroinflammation?
5 answers
Allergies have been associated with neuroinflammation through the involvement of mast cells (MCs). MCs, known for their role in allergic reactions, can contribute to neuroinflammation by releasing inflammatory mediators, affecting the blood-brain barrier (BBB) integrity, and interacting with glial cells and neurons. Studies suggest that MC activation may play a part in the pathogenesis of neurodevelopmental disorders like attention deficit hyperactivity disorder (ADHD). Furthermore, MCs have been implicated in exacerbating CNS damage in conditions such as brain ischemia and hemorrhage, while also potentially playing a protective role in traumatic brain injury. The intricate relationship between allergies, MC activation, and neuroinflammation underscores the complex interplay between the immune system and the central nervous system in various pathological processes.
What is then the most common cell line that OECT interact with?
9 answers
The question regarding the most common cell line that Organic Electrochemical Transistors (OECT) interact with cannot be directly answered from the provided contexts, as none of the abstracts specifically mention Organic Electrochemical Transistors (OECT) or their interactions with cell lines. However, by analyzing the information given in the contexts, we can infer the types of cell lines and cellular interactions that are prevalent in research, which might provide insight into cell lines that could potentially interact with OECTs in hypothetical scenarios. From the contexts provided, several cell lines and their interactions are highlighted, focusing on cancer research, immune responses, and cell-cell interactions in various environments. For instance, the Ehrlich ascites tumor cell lines and the P388 murine leukemia cell line are discussed in terms of their growth characteristics and immunogenic interactions in immune-competent mice. Similarly, the interaction between mesenchymal stem cells (MSC) and ovarian cancer cells demonstrates the influence of co-culturing on cell growth and expression of specific markers. Additionally, the Chinese hamster ovary (CHO) cell line is mentioned for its use in the production of recombinant human proteins and the formation of cell aggregates. Given the diversity of cell lines and their specific interactions detailed in the contexts, it's challenging to pinpoint a single cell line as the most common for interaction with OECTs without direct references to such technology. However, the emphasis on cancer cell lines (e.g., Ehrlich ascites tumor cells, P388 murine leukemia, and ovarian cancer cells) and their interactions with the immune system or in co-culture systems suggests that these cell lines are of significant interest in biomedical research. Therefore, in the realm of research focusing on cellular interactions and responses, cancer cell lines and MSCs might be considered among the most relevant for potential interactions with OECTs, given their prominence in the study of cell behavior, drug screening, and therapeutic applications.
What's the role of IL-33/ST2 axis in prostate cancer?
4 answers
The IL-33/ST2 axis plays a crucial role in cancer immunity and immunotherapy. IL-33, upon release, binds to its receptor ST2, influencing immune responses in the tumor microenvironment (TME). In prostate cancer, the IL-33/ST2 axis affects macrophage polarization, promoting tumor growth by altering macrophage metabolism from OXPHOS to glycolysis. Furthermore, IL-33, through ST2, modulates immune responses in the TME, impacting both innate and adaptive immune cells during cancer initiation and progression. Studies also indicate that IL-33 levels differ in cardiovascular disease patients, suggesting a potential role in differentiating disease severity and outcomes, emphasizing the need for further research in this area. Overall, the IL-33/ST2 axis emerges as a potential therapeutic target for cancer immunotherapy, including prostate cancer.
How does IL-34 contribute to angiogenesis of prostate cancer?
4 answers
IL-34 plays a significant role in promoting angiogenesis in prostate cancer (PCA) by influencing the tumor microenvironment. Endothelial cells, which express CD31 and CD34, are increased in PCA and contribute to metastasis by enhancing cancer cell invasion. IL-34, secreted by various cells including tumor cells, has been implicated in promoting cancer growth and metastasis in various cancers. Additionally, IL-34 is involved in regulating immune cells, particularly macrophages, in the tumor microenvironment, which can lead to immune suppression and cancer progression. The combination of IL-34 inhibition with immune checkpoint inhibitors (ICIs) has been proposed as a potential strategy to overcome resistance to ICIs and improve cancer treatment efficacy. Therefore, targeting IL-34 may offer a promising approach to hinder angiogenesis and enhance therapeutic outcomes in prostate cancer.
What's the role of IL-33 in prostate cancer?
4 answers
IL-33 plays a significant role in prostate cancer by influencing its development and progression. Research indicates that IL-33 levels are elevated in prostate cancer patients, especially in advanced stages, suggesting a potential contribution to disease progression. IL-33 has been found to affect the expression of various cytokines that promote tumor growth, angiogenesis, and immune cell recruitment, thereby promoting tumorigenesis. Additionally, genetic alterations in the IL-33 gene, such as the rs1929992 SNP, have been associated with an increased risk of prostate cancer development. These findings highlight the dual nature of IL-33 in cancer, where it can act both pro-tumor and anti-tumor, emphasizing the complexity of its role in prostate cancer pathogenesis.
Where is ACE2 expressed?
5 answers
ACE2 is expressed in various locations within the human body. In Alzheimer's disease, ACE2 is mainly found on the surface of brain endothelial cells. Human spermatozoa express ACE2 uniformly on the sperm head and flagellum, with the short-ACE2 isoform concentrated in the post-acrosomal region and midpiece. Additionally, ACE2 is present in different tissues, with unique regulation and expression levels in mice and humans, influencing susceptibility to SARS-CoV-2 infection and the subsequent host response. The expression of ACE2 in spermatozoa and its potential role in male reproduction warrant further investigation to understand its implications fully.
Why the ros of erlotininb nanoparticles are higher than free erlotininb?
5 answers
The redox-responsive and pH-sensitive nanoparticle drug delivery systems designed for erlotinib encapsulation showed higher cytotoxicity due to their ability to release the drug in response to ROS. Similarly, ROS-sensitive nanoparticles loaded with paclitaxel exhibited enhanced anti-tumor efficacy compared to free paclitaxel or nab-paclitaxel. These nanoparticles utilize ROS-triggered degradation mechanisms to release the drug payload effectively in the tumor microenvironment, leading to improved therapeutic outcomes. Additionally, the ROS-responsive, FcRn-targeting, and caged nanoparticles carrying Fedratinib demonstrated prolonged residence time and enhanced therapeutic effects in treating eosinophilic asthma. Therefore, the utilization of ROS-responsive nanoparticles enhances drug delivery efficiency and efficacy, making them more potent than free drugs in combating cancer and other diseases.
What specific biomarkers have been identified as predictors of treatment response in bladder cancer patients?
10 answers
In the quest to enhance treatment outcomes for bladder cancer (BC) patients, several biomarkers have emerged as potential predictors of treatment response. A chemotherapy response type gene (CRTG) signature, comprising 9 genes, has been established to predict chemotherapy response, with Rac family small GTPase 3 (RAC3) identified as a key biomarker for chemoresistance, influencing the PAK1-ERK1/2 pathway. Additionally, the molecular landscape of bladder cancer has revealed genetic alterations and molecular subtypes, suggesting a shift towards personalized treatment, although specific biomarkers were not detailed. Exosome-related genes have also been spotlighted, with a prognostic model developed from differentially expressed genes (DEGs) related to exosomes, indicating their significance in BC patient survival and treatment outcome prediction. The Neutrophil-to-Lymphocyte Ratio (NLR) has been hypothesized as a predictive marker for immune checkpoint inhibitor (ICI) therapy response, with lower NLR associated with longer event-free survival. Despite the absence of clinically approved predictive biomarkers, DNA damage repair (DDR) gene alterations and molecular subtypes have shown promise in guiding therapy, pending validation from clinical trials. Systemic inflammatory response (SIR) markers, particularly the neutrophil-to-lymphocyte ratio (NLR) and monocyte-to-lymphocyte ratio (MLR), have been identified as independent predictive factors for recurrence-free survival (RFS) and progression-free survival (PFS) in non-muscle invasive bladder cancer (NMIBC). Tumor microenvironment (TME)-related genes have been associated with prognosis and immunotherapy response, offering a new avenue for predicting treatment outcomes. Biodynamic signatures, reflecting the phenomic biomarkers within a tumor, have shown high predictive accuracy for neoadjuvant chemotherapy (NAC) response in muscle-invasive bladder cancer (MIBC) patients. Various tissue-based, urine-based, and serum-based biomarkers across non–muscle-invasive and muscle-invasive BC have been evaluated for their predictive capabilities, although rigorous validation remains a challenge. Lastly, molecular signatures, including mRNA and miRNA differential expression profiles, have been explored to differentiate NAC responders from non-responders, highlighting the potential of molecular pathways and subtype classifications in predicting chemotherapy response.
How to enhance brain permeability of small molecules?
5 answers
To enhance brain permeability of small molecules, various strategies can be employed. Medicinal chemists focus on modifying small molecules to improve diffusion, reduce efflux, and activate carrier transporters, facilitating their passage through the blood-brain barrier (BBB). Utilizing native nutrient transporters of the BBB is another approach to increase CNS drug delivery by exploiting these transporters to carry drugs through endothelial cells. Computational prediction models based on physicochemical properties and molecular substructures can aid in understanding the mechanisms contributing to BBB permeability, guiding the identification of BBB-permeable compounds by matching their properties to transport mechanisms. Additionally, microfluidic-based platforms have been developed to model and predict brain tissue uptake of small molecules, providing insights into BBB permeability and enabling visualization of transcytosis processes.
Who do PTEN may be served as a biomarker of resistance to anti-PD-1 immunotherapy in lung adenocarcinoma?
5 answers
PTEN mutation may serve as a biomarker of resistance to anti-PD-1 immunotherapy in lung adenocarcinoma. Studies have shown that patients with low PTEN levels in their tumors exhibit higher levels of immunosuppressive factors like CXCL10, PD-L1, and PD-L2, leading to worse outcomes when treated with immunotherapy. Additionally, PTEN loss in lung squamous cell carcinoma (LUSC) creates an immunosuppressive tumor microenvironment by promoting the infiltration of regulatory T cells (Tregs) and expressing higher levels of immunosuppressive genes, resulting in resistance to anti-PD-1 therapy. This resistance can be reversed therapeutically by targeting the immunosuppressive microenvironment through treatments like TLR agonists and anti-TGF-β antibodies, leading to tumor rejection and immunological memory.
What makes HNSCC cells form round clusters when cultured on Matrigel?
4 answers
When HNSCC cells are cultured on Matrigel, they form round clusters due to the influence of specific genetic and functional characteristics. Studies have shown that single cell-derived spheroids from HNSCC cell lines exhibit distinct clusters with unique gene expression signatures enriched in pathways like cell cycle, anti-apoptosis, and pro-survival genes associated with EGFR, Wnt, and Notch signaling pathways. Additionally, the formation of spheroids in a 3D culture system can induce the differentiation of cells towards specific phenotypes, as seen in the case of meibomian gland epithelial cells where 3D spheroid culture facilitated meibocyte differentiation. Furthermore, the molecular mechanisms regulating the formation of spheroid cell clusters involve signaling pathways like focal adhesion kinase (FAK) signaling, highlighting the importance of specific protein phosphorylation for spheroid formation.