Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Cell Res ; 439(1): 114073, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38704079

RESUMO

Determining the appropriate source of antigens for optimal antigen presentation to T cells is a major challenge in designing dendritic cell (DC) -based therapeutic strategies against hepatocellular carcinoma (HCC). Tumor-derived exosomes (Tex) express a wide range of tumor antigens, making them a promising source of antigens for DC vaccines. As reported, the exosomes secreted by tumor cells can inhibit the antitumor function of immune cells. In this study, we transfected hepatocellular carcinoma cells with Rab27a to enhance the yield of exosomes, which were characterized using transmission electron microscopy and Western blot analysis. We found that Tex secreted by overexpressing Rab27a Hepatocellular carcinoma cell lines pulsed DC is beneficial for the differentiation and maturation of DCs but inhibits the secretion of the IL-12 cytokine. Consequently, we developed a complementary immunotherapy approach by using Tex as an antigen loaded onto DCs, in combination with the cytokine IL-12 to induce antigen-specific cytotoxic T lymphocytes (CTLs). The results indicated that the combination of DC-Tex and IL-12 was more effective in stimulating T lymphocyte proliferation, releasing IFN-γ, and enhancing cytotoxicity compared to using exosomes or IL-12 alone. Additionally, the inclusion of IL-12 also compensated for the reduced IL-2 secretion by DCs caused by Tex. Moreover, in a BALB/c nude mice model of hepatocellular carcinoma, CTLs induced by DC-Tex combined with IL-12 maximized the tumor-specific T-cell immune effect and suppressed tumor growth. Thus, Tex provides a novel and promising source of antigens, with cytokines compensating for the shortcomings of Tex as a tumor antigen. This work helps to clarify the role of exosomes in tumor immunotherapy and may offer a safe and effective prospective strategy for the clinical application of exosome-based cellular immunotherapy.


Assuntos
Carcinoma Hepatocelular , Células Dendríticas , Exossomos , Interleucina-12 , Neoplasias Hepáticas , Proteínas rab27 de Ligação ao GTP , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Exossomos/metabolismo , Animais , Interleucina-12/metabolismo , Interleucina-12/genética , Proteínas rab27 de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP/genética , Carcinoma Hepatocelular/terapia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/genética , Camundongos , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/genética , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Camundongos Endogâmicos BALB C , Imunoterapia/métodos
2.
Breast Cancer Res ; 26(1): 57, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553754

RESUMO

Breast cancer continues to pose a substantial worldwide health concern, demanding a thorough comprehension of the complex interaction between cancerous cells and the immune system. Recent studies have shown the significant function of exosomes in facilitating intercellular communication and their participation in the advancement of cancer. Tumor-derived exosomes have been identified as significant regulators in the context of breast cancer, playing a crucial role in modulating immune cell activity and contributing to the advancement of the illness. This study aims to investigate the many effects of tumor-derived exosomes on immune cells in the setting of breast cancer. Specifically, we will examine their role in influencing immune cell polarization, facilitating immunological evasion, and modifying the tumor microenvironment. Furthermore, we explore the nascent domain of exosomes produced from immune cells and their prospective involvement in the prevention of breast cancer. This paper focuses on new research that emphasizes the immunomodulatory characteristics of exosomes produced from immune cells. It also explores the possibility of these exosomes as therapeutic agents or biomarkers for the early identification and prevention of breast cancer. The exploration of the reciprocal connections between exosomes formed from tumors and immune cells, together with the rising significance of exosomes derived from immune cells, presents a potential avenue for the advancement of novel approaches in the field of breast cancer therapy and prevention.


Assuntos
Neoplasias da Mama , Exossomos , Neoplasias , Humanos , Feminino , Neoplasias da Mama/patologia , Exossomos/patologia , Estudos Prospectivos , Comunicação Celular , Microambiente Tumoral
3.
Int J Cancer ; 154(12): 2031-2042, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38500385

RESUMO

Tumor-associated myeloid cells (TAMCs) play a crucial role in orchestrating the dynamics of the tumor immune microenvironment. This heterogeneous population encompasses myeloid-derived suppressor cells, tumor-associated macrophages and dendritic cells, all of which contribute to the establishment of an immunosuppressive milieu that fosters tumor progression. Tumor-derived exosomes (TEXs), small extracellular vesicles secreted by tumor cells, have emerged as central mediators in intercellular communication within the tumor microenvironment. In this comprehensive review, we explore the intricate mechanisms through which TEXs modulate immune-suppressive effects on TAMCs and their profound implications in cancer progression. We delve into the multifaceted ways in which TEXs influence TAMC functions, subsequently affecting tumor immune evasion. Furthermore, we elucidate various therapeutic strategies aimed at targeting TEX-mediated immune suppression, with the ultimate goal of bolstering antitumor immunity.


Assuntos
Exossomos , Células Supressoras Mieloides , Neoplasias , Humanos , Exossomos/patologia , Neoplasias/patologia , Terapia de Imunossupressão , Células Mieloides , Microambiente Tumoral
4.
Small ; 20(30): e2308335, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38420895

RESUMO

Tumor-derived exosomes (TDEs) induced extracellular microenvironment has recently been validated to be critical for tumor progression and metastasis, however, remodeling it for oncotherapy still remains a major challenge due to difficulty in regulation of TDEs secretion. Herein, the supramolecular chiral nanofibers, composed of L/D-phenylalanine derivates (L/D-Phe) and linear hyaluronic acid (HA), are successfully employed to construct TDEs induced anti-tumor extracellular microenvironment. The left-handed L-Phe @HA nanofibers significantly inhibit TDEs secretion into extracellular microenvironment, which results in suppression of tumor proliferation and metastasis in vitro and vivo. Biological assays and theoretical modeling reveal that these results are mainly attributed to strong adsorption of the key exosomes transporters (Ras-related protein Rab-27A and synaptosome-associated protein 23) on left-handed L-Phe @HA nanofibers via enhanced stereoselective interaction, leading to degradation and phosphorylated dropping of exosomes transporters. Subsequently, transfer function of exosomes transporters is limited, which causes remarkable inhibition of TDEs secretion. These findings provide a promising novel insight of chiral functional materials to establish an anti-tumor extracellular microenvironment via regulation of TDEs secretion.


Assuntos
Exossomos , Nanofibras , Microambiente Tumoral , Nanofibras/química , Exossomos/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Humanos , Linhagem Celular Tumoral , Animais , Ácido Hialurônico/química , Proliferação de Células/efeitos dos fármacos
5.
Chin J Cancer Res ; 36(2): 151-166, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38751437

RESUMO

Throughout tumorigenesis, the co-evolution of tumor cells and their surrounding microenvironment leads to the development of malignant phenotypes. Cellular communication within the tumor microenvironment (TME) plays a critical role in influencing various aspects of tumor progression, including invasion and metastasis. The release of exosomes, a type of extracellular vesicle, by most cell types in the body, is an essential mediator of intercellular communication. A growing body of research indicates that tumor-derived exosomes (TDEs) significantly expedite tumor progression through multiple mechanisms, inducing epithelial-mesenchymal transition and macrophage polarization, enhancing angiogenesis, and aiding in the immune evasion of tumor cells. Herein, we describe the formation and characteristics of the TME, and summarize the contents of TDEs and their diverse functions in modulating tumor development. Furthermore, we explore potential applications of TDEs in tumor diagnosis and treatment.

6.
Med Res Rev ; 43(4): 1141-1200, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36929669

RESUMO

Epithelial-mesenchymal transition (EMT) is a complex process with a primordial role in cellular transformation whereby an epithelial cell transforms and acquires a mesenchymal phenotype. This transformation plays a pivotal role in tumor progression and self-renewal, and exacerbates resistance to apoptosis and chemotherapy. EMT can be initiated and promoted by deregulated oncogenic signaling pathways, hypoxia, and cells in the tumor microenvironment, resulting in a loss-of-epithelial cell polarity, cell-cell adhesion, and enhanced invasive/migratory properties. Numerous transcriptional regulators, such as Snail, Slug, Twist, and ZEB1/ZEB2 induce EMT through the downregulation of epithelial markers and gain-of-expression of the mesenchymal markers. Additionally, signaling cascades such as Wnt/ß-catenin, Notch, Sonic hedgehog, nuclear factor kappa B, receptor tyrosine kinases, PI3K/AKT/mTOR, Hippo, and transforming growth factor-ß pathways regulate EMT whereas they are often deregulated in cancers leading to aberrant EMT. Furthermore, noncoding RNAs, tumor-derived exosomes, and epigenetic alterations are also involved in the modulation of EMT. Therefore, the regulation of EMT is a vital strategy to control the aggressive metastatic characteristics of tumor cells. Despite the vast amount of preclinical data on EMT in cancer progression, there is a lack of clinical translation at the therapeutic level. In this review, we have discussed thoroughly the role of the aforementioned transcription factors, noncoding RNAs (microRNAs, long noncoding RNA, circular RNA), signaling pathways, epigenetic modifications, and tumor-derived exosomes in the regulation of EMT in cancers. We have also emphasized the contribution of EMT to drug resistance and possible therapeutic interventions using plant-derived natural products, their semi-synthetic derivatives, and nano-formulations that are described as promising EMT blockers.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias , Humanos , Transição Epitelial-Mesenquimal/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Hedgehog/metabolismo , Neoplasias/metabolismo , Fatores de Transcrição , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral
7.
Am J Physiol Cell Physiol ; 324(6): C1307-C1319, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37154491

RESUMO

It is a widespread and difficult problem that refractory diabetic wounds have a poor local environment and prolonged inflammatory irritation. Tumor cell-derived exosomes play an important role in the development of tumors, as they can promote tumor cell proliferation, migration, and invasion and enhance tumor cell activity. However, tumor tissue-derived exosomes (Ti-Exos) have been less studied, and it is unclear how they affect wound healing. In this study, we extracted Ti-Exos from human oral squamous carcinoma and paracancerous tissue by ultracentrifugation, size exclusion chromatography, and ultrafiltration and performed exosome characterization. In vitro, the oral squamous cell carcinoma tissue-derived exosomes (OSCC Ti-Exos) promoted the proliferation and migration of endothelial cells, keratinocytes, and fibroblasts. In addition, in vivo experiments showed that the OSCC Ti-Exos accelerated the healing of diabetic wounds and were safe in mice. In contrast, there was no promoting effect of paracancerous tissue-derived exosomes either in vivo or in vitro. In conclusion, OSCC Ti-Exos promoted the healing of diabetic wounds, demonstrated preliminary biosafety in mice, and have promise as therapeutic applications.NEW & NOTEWORTHY Diabetic wound healing has become a public health issue that lacks effective treatment. We collected oral squamous cell carcinoma samples and paracancerous tissue and extracted Ti-Exos for verification. In vitro assays revealed that OSCC Ti-EVs could enhance the proliferation and migration of endothelial cells, keratinocytes, and fibroblasts in diabetic cell model. In vivo assays also verified that OSCC Ti-Exos could promote diabetic wound healing, demonstrated preliminary biosafety in mice, and have promise as therapeutic applications.


Assuntos
Carcinoma de Células Escamosas , Diabetes Mellitus , Exossomos , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Camundongos , Animais , Células Endoteliais , Carcinoma de Células Escamosas de Cabeça e Pescoço , Exossomos/química , Cicatrização , Proliferação de Células
8.
Cell Commun Signal ; 21(1): 150, 2023 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-37349803

RESUMO

Multiple studies have shown that extracellular vesicles (EVs) play a key role in the process of information transfer and material transport between cells. EVs are classified into different types according to their sizes, which includes the class of exosomes. In comparison to normal EVs, tumor-derived EVs (TDEs) have both altered components and quantities of contents. TDEs have been shown to help facilitate an environment conducive to the occurrence and development of tumor by regulation of glucose, lipids and amino acids. Furthermore, TDEs can also affect the host metabolism and immune system. EVs have been shown to have multiple clinically useful properties, including the use of TDEs as biomarkers for the early diagnosis of diseases and using the transport properties of exosomes for drug delivery. Targeting the key bioactive cargoes of exosomes could be applied to provide new strategies for the treatment of tumors. In this review, we summarize the finding of studies focused on measuring the effects of TDE on tumor-related microenvironment and systemic metabolism. Video Abstract.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Humanos , Vesículas Extracelulares/metabolismo , Exossomos/metabolismo , Neoplasias/patologia , Comunicação Celular , Biomarcadores/metabolismo , Microambiente Tumoral
9.
Cell Biol Toxicol ; 39(2): 519-536, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35674868

RESUMO

It has been manifested that tumor-derived exosomes (Exos) can deliver long noncoding RNAs to participate in gastric cancer (GC) progression. In this research, we intended to dissect out whether tumor-derived Exos carried LINC01091 to afflict the growth and metastasis of GC. GC tissues and human GC cells were attained for RNA and protein quantification. Accordingly, LINC01091, ELF4, and CDX2 were abundant but microRNA (miR)-128-3p was underexpressed in GC tissues and cells. Exos were isolated from LINC01091-silenced GC cells (Exo-sh-LINC01091). GC cells were co-cultured with Exo-sh-LINC01091 or manipulated with miR mimic, inhibitor, or overexpressing or silencing plasmids. Exo-sh-LINC01091, LINC01091, ELF4 or CDX2 silencing, or miR-128-3p upregulation augmented GC cell proliferative, migrating, and invasive properties. In addition, luciferase, RNA pull-down, and ChIP assays offered evidence supporting the mechanism that LINC01091 bound to miR-128-3p that inversely targeted ELF4, and ELF4 transcriptionally activated CDX2 by binding to its promoter in GC cells. Moreover, Exo-sh-LINC01091 modulated the miR-128-3p/ELF4/CDX2 axis and restrained the tumorigenesis and metastasis in vivo. Conclusively, LINC01091 shuttled by tumor-derived Exos might expedite GC development by activating the ELF4/CDX2 axis via miR-128-3p downregulation.


Assuntos
Exossomos , MicroRNAs , Neoplasias Gástricas , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias Gástricas/metabolismo , Exossomos/genética , Exossomos/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Transcrição CDX2/genética , Fator de Transcrição CDX2/metabolismo
10.
Biotechnol Appl Biochem ; 70(5): 1754-1771, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37254633

RESUMO

M2 macrophages are the most prevalent type in the tumor microenvironment and their polarization to M1 type can be used as a potential cancer immunotherapy. Here, we investigated the role of tumor microenvironment and particularly purified exosomes in M2 to M1 macrophage polarization. Rapamycin treatment on triple-negative breast cancer cells (TNBC) was performed. Tumor cells-derived exosomes (called texosomes) were isolated and characterized using scanning electron microscopy, transmission electron microscopy, dynamic light scattering, high-performance liquid chromatography, Fourier transform infrared, and Western blot assays. M2 mouse peritoneal macrophages were treated with rapamycin or rapamycin-texosome. Then, M1/M2 phenotype-specific marker genes and proteins were measured to assess the degree of M2 to M1 polarization. Finally, nitric oxide (NO) production, phagocytosis, and efferocytosis assays were assessed to verify the functionality of the polarized macrophages. Purified rapamycin-texosomes significantly increased the expression of the M1 markers (Irf5, Nos2, and CD86) and decreased M2 markers (Arg, Ym1, and CD206). In addition, the levels of M1-specific cytokines tumor necrosis factor alpha and interleukin 1ß (IL-1ß) were increased, whereas the levels of M2 specific cytokines IL-10 and transforming growth factor beta were declined. Furthermore, texosome treatment increased NO concentration and phagocytosis and decreased efferocytosis indicating M1 polarization. These findings suggest rapamycin-texosomes can induce M2 to M1 macrophages polarization as a potential immunotherapy for TNBC.


Assuntos
Exossomos , Neoplasias de Mama Triplo Negativas , Humanos , Camundongos , Animais , Sirolimo , Exossomos/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Macrófagos/metabolismo , Citocinas/metabolismo , Fenótipo , Microambiente Tumoral , Fatores Reguladores de Interferon/metabolismo
11.
Cell Mol Life Sci ; 79(11): 572, 2022 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-36308630

RESUMO

Almost all clinical oncologists agree that the discovery of reliable, accessible, and non-invasive biomarkers is necessary to decrease cancer mortality. It is possible to employ reliable biomarkers to diagnose cancer in the early stages, predict the patient prognosis, follow up the response to treatment, and estimate the risk of disease recurrence with high sensitivity and specificity. Extracellular vesicles (EVs), especially exosomes, have been the focus of translational research to develop such biomarkers over the past decade. The abundance and distribution of exosomes in bodily fluids, including serum, saliva, and urine, as well as their ability to transport various biomolecules (nucleic acids, proteins, and lipids) derived from their parent cells, make exosomes reliable, accessible, and potent biomarkers for diagnosis and follow-up of solid and hematopoietic tumors. In addition, exosomes play a vital role in various cellular processes, including tumor progression, by participating in intercellular communication. Although these advantages underline the high potential of tumor-derived exosomes as diagnostic biomarkers, the lack of standardized effective methods for their isolation, identification, and precise characterization makes their application challenging in clinical settings. We discuss the importance of non-coding RNAs (ncRNAs) in cellular processes, and the role of tumor-derived exosomes containing ncRNAs as potential biomarkers in several types of cancer. In addition, the advantages and challenges of these studies for translation into clinical applications are covered.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/tratamento farmacológico , RNA não Traduzido/genética , RNA não Traduzido/metabolismo , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Biomarcadores/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo
12.
Int J Mol Sci ; 25(1)2023 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-38203424

RESUMO

Exosomes, a type of extracellular vesicle with a diameter of approximately 100 nm that is secreted by all cells, regulate the phenotype and function of recipient cells by carrying molecules such as proteins, nucleic acids, and lipids and are important mediators of intercellular communication. Exosomes are involved in various physiological and pathological processes such as immunomodulation, angiogenesis, tumorigenesis, metastasis, and chemoresistance. Due to their excellent properties, exosomes have shown their potential application in the clinical diagnosis and treatment of disease. The functions of exosomes depend on their biogenesis, uptake, and composition. Thus, a deeper understanding of these processes and regulatory mechanisms can help to find new targets for disease diagnosis and therapy. Therefore, this review summarizes and integrates the recent advances in the regulatory mechanisms of the entire biological process of exosomes, starting from the formation of early-sorting endosomes (ESCs) by plasma membrane invagination to the release of exosomes by fusion of multivesicular bodies (MVBs) with the plasma membrane, as well as the regulatory process of the interactions between exosomes and recipient cells. We also describe and discuss the regulatory mechanisms of exosome production in tumor cells and the potential of exosomes used in cancer diagnosis and therapy.


Assuntos
Exossomos , Vesículas Extracelulares , Endossomos , Vesículas Transportadoras , Transporte Biológico
13.
J Cell Physiol ; 237(3): 1648-1660, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34825383

RESUMO

Cancer is a gravely important health issue all over the world and has been spreading fast. In recent years immune checkpoint treatment options have been used extensively as a primary line of treatment for different cancer types. PD-1 and its ligand, PD-L1, are members of the immune-checkpoints superfamily. Anti-PD-L1 and anti-PD-1 antibodies have shown efficacy against different cancer types, but fewer than 30% of patients have shown robust therapeutic responses and, therefore, it is hypothesized that exosomal PD-L1 is the mechanism to blame for failure in primary immune checkpoint therapy. The identical membrane topology of exosomal PD-L1 with tumor cell membrane-type provides the possibility to mimic immunosuppressive effects of tumor cell membrane PD-L1. In this review, it is discussed whether exosomal PD-L1 binds to antibodies and hence resistance to immunotherapy will be developed, and targeting exosome biogenesis inhibition can provide a new strategy to overcome tumor resistance to anti-PD-L1 therapy. Diagnostic and prognostic values of exosomal PD-L1 in different cancer types are discussed. Multiple clinical studies conclude that the level of tumor-derived exosomes (TEXs) as a biomarker for diagnosis could distinguish cancer patients from healthy controls. Elevated exosomal PD-L1 levels may be predictive of advanced disease stages, cancer metastasis, lower response to anti-PD-1/PD-L1 therapy, lower overall survival rates, and poor tumor prognosis. These novel findings of TEXs serve as promising therapeutic targets for early diagnosis and prevention of cancer progression.


Assuntos
Exossomos , Neoplasias , Antígeno B7-H1/metabolismo , Exossomos/metabolismo , Humanos , Tolerância Imunológica , Fatores Imunológicos/farmacologia , Imunoterapia , Neoplasias/patologia
14.
Mol Cancer ; 21(1): 45, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35148751

RESUMO

BACKGROUND: Dendritic cells (DCs) are central for the initiation and regulation of innate and adaptive immunity in the tumor microenvironment. As such, many kinds of DC-targeted vaccines have been developed to improve cancer immunotherapy in numerous clinical trials. Targeted delivery of antigens and adjuvants to DCs in vivo represents an important approach for the development of DC vaccines. However, nonspecific activation of systemic DCs and the preparation of optimal immunodominant tumor antigens still represent major challenges. METHODS: We loaded the immunogenic cell death (ICD) inducers human neutrophil elastase (ELANE) and Hiltonol (TLR3 agonist) into α-lactalbumin (α-LA)-engineered breast cancer-derived exosomes to form an in situ DC vaccine (HELA-Exos). HELA-Exos were identified by transmission electron microscopy, nanoscale flow cytometry, and Western blot analysis. The targeting, killing, and immune activation effects of HELA-Exos were evaluated in vitro. The tumor suppressor and immune-activating effects of HELA-Exos were explored in immunocompetent mice and patient-derived organoids. RESULTS: HELA-Exos possessed a profound ability to specifically induce ICD in breast cancer cells. Adequate exposure to tumor antigens and Hiltonol following HELA-Exo-induced ICD of cancer cells activated type one conventional DCs (cDC1s) in situ and cross-primed tumor-reactive CD8+ T cell responses, leading to potent tumor inhibition in a poorly immunogenic triple negative breast cancer (TNBC) mouse xenograft model and patient-derived tumor organoids. CONCLUSIONS: HELA-Exos exhibit potent antitumor activity in both a mouse model and human breast cancer organoids by promoting the activation of cDC1s in situ and thus improving the subsequent tumor-reactive CD8+ T cell responses. The strategy proposed here is promising for generating an in situ DC-primed vaccine and can be extended to various types of cancers. Scheme 1. Schematic illustration of HELA-Exos as an in situ DC-primed vaccine for breast cancer. (A) Allogenic breast cancer-derived exosomes isolated from MDA-MB-231 cells were genetically engineered to overexpress α-LA and simultaneously loaded with the ICD inducers ELANE and Hiltonol (TLR3 agonist) to generate HELA-Exos. (B) Mechanism by which HELA-Exos activate DCs in situ in a mouse xenograft model ofTNBC. HELA-Exos specifically homed to the TME and induced ICD in cancer cells, which resulted in the increased release of tumor antigens, Hiltonol, and DAMPs, as well as the uptake of dying tumor cells by cDC1s. The activated cDC1s then cross-primed tumor-reactive CD8+ T cell responses. (C) HELA-Exos activated DCs in situ in the breast cancer patient PBMC-autologous tumor organoid coculture system. ABBREVIATIONS: DCs: dendritic cells; α-LA: α-lactalbumin; HELA-Exos: Hiltonol-ELANE-α-LA-engineered exosomes; ICD: immunogenic cell death; ELANE: human neutrophil elastase; TLR3: Toll-like receptor 3; TNBC: triple-negative breast cancer; TME: tumor microenvironment; DAMPs: damage-associated molecular patterns; cDC1s: type 1 conventional dendritic cells; PBMCs: peripheral blood mononuclear cells.


Assuntos
Neoplasias da Mama , Vacinas Anticâncer , Exossomos , Vacinas , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Células Dendríticas , Feminino , Humanos , Leucócitos Mononucleares , Camundongos , Microambiente Tumoral , Vacinas/metabolismo
15.
Clin Proteomics ; 19(1): 2, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34996345

RESUMO

BACKGROUND: Early detection of small cell lung cancer (SCLC) crucially demands highly reliable markers. Growing evidence suggests that extracellular vesicles carry tumor cell-specific cargo suitable as protein markers in cancer. Quantitative proteomic profiling of circulating microvesicles and exosomes can be a high-throughput platform for discovery of novel molecular insights and putative markers. Hence, this study aimed to investigate proteome dynamics of plasma-derived microvesicles and exosomes in newly diagnosed SCLC patients to improve early detection. METHODS: Plasma-derived microvesicles and exosomes from 24 healthy controls and 24 SCLC patients were isolated from plasma by either high-speed- or ultracentrifugation. Proteins derived from these extracellular vesicles were quantified using label-free mass spectrometry and statistical analysis was carried out aiming at identifying significantly altered protein expressions between SCLC patients and healthy controls. Furthermore, significantly expressed proteins were subjected to functional enrichment analysis to identify biological pathways implicated in SCLC pathogenesis. RESULTS: Based on fold change (FC) ≥ 2 or ≤ 0.5 and AUC ≥ 0.70 (p < 0.05), we identified 10 common and 16 and 17 unique proteins for microvesicles and exosomes, respectively. Among these proteins, we found dysregulation of coagulation factor XIII A (Log2 FC = - 1.1, p = 0.0003, AUC = 0.82, 95% CI: 0.69-0.96) and complement factor H-related protein 4 (Log2 FC = 1.2, p = 0.0005, AUC = 0.82, 95% CI; 0.67-0.97) in SCLC patients compared to healthy individuals. Our data may indicate a novel tumor-suppressing role of blood coagulation and involvement of complement activation in SCLC pathogenesis. CONCLUSIONS: In comparing SCLC patients and healthy individuals, several differentially expressed proteins were identified. This is the first study showing that circulating extracellular vesicles may encompass specific proteins with potential diagnostic attributes for SCLC, thereby opening new opportunities as novel non-invasive markers.

16.
Nanotechnology ; 33(29)2022 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-35051909

RESUMO

The physical properties of tumor-derived exosomes have gained much attention because they are helpful to better understand the exosomes in biomedicine. In this study, the conductive atomic force microscopy (C-AFM) was employed to perform the electrical characterizations of exosomes, and it obtained the topography and current images of samples simultaneously. The exosomes were absorbed onto the mica substrates coated with a gold film of 20 nm thick for obtaining the current images of samples by C-AFM in air. The results showed that the single exosomes had the weak conductivity. Furthermore, the currents on exosomes were measured at different bias voltages and pH conditions. It illustrated that the conductivity of exosomes was affected by external factors such as bias voltages and solutions with different pH values. In addition, the electrical responses of low and high metastatic potential cell-derived exosomes were also compared under different voltages and pH conditions. This work is important for better understanding the physical properties of tumor-derived exosomes and promoting the clinical applications of tumor-derived exosomes.


Assuntos
Exossomos , Neoplasias , Condutividade Elétrica , Eletricidade , Exossomos/química , Humanos , Microscopia de Força Atômica/métodos
17.
Cell Biochem Funct ; 40(3): 248-262, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35285964

RESUMO

Most eukaryotic cells secrete extracellular vesicles (EVs), which contribute to intracellular communication through transferring different biomolecules such as proteins, RNAs, and lipids to cells. Two main types of EVs are exosomes and microvesicles. Exosomes originate from multivesicular bodies, while microvesicles are shed from the plasma membrane. Mechanisms of exosomes and microvesicle biogenesis/trafficking are complex and many molecules are involved in their biogenesis and secretion. Tumor-derived EVs contain oncogenic molecules that promote tumor growth, metastasis, immune surveillance, angiogenesis, and chemoresistance. A growing body of evidence indicates various compounds can inhibit biogenesis and secretion of EVs from cells and several experiments were conducted to use EVs-inhibitors for understanding the biology of the cells or for understanding the pathology of several diseases like cancer. However, the nontargeting effects of drugs/inhibitors remain a concern. Our current knowledge of EVs biogenesis and their inhibition from tumor cells may provide an avenue for cancer management. In this review, we shed light on exosomes and microvesicles biogenesis, key roles of tumor-derived EVs, and discuss methods used to inhibition of EVs by different inhibitors.


Assuntos
Exossomos , Vesículas Extracelulares , Neoplasias , Carcinogênese/metabolismo , Membrana Celular/metabolismo , Exossomos/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Neoplasias/metabolismo
18.
Semin Immunol ; 35: 69-79, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29289420

RESUMO

Mesenchymal stem cells (MSCs) are a major component of the tumor microenvironment (TME) and play a key role in promoting tumor progression. The tumor uses exosomes to co-opt MSCs and re-program their functional profile from normally trophic to pro-tumorigenic. These tumor-derived small vesicles called "TEX" carry and deliver a cargo rich in proteins and nucleic acids to MSCs. Upon interactions with surface receptors on MSCs and uptake of the exosome cargo by MSCs, molecular, transcriptional and translational changes occur that convert MSCs into producers of factors that are necessary for tumor growth and that also alter functions of non-tumor cells in the TME. The MSCs re-programmed by TEX become avid producers of their own exosomes that carry and deliver mRNA and miRNA species as well as molecular signals not only back to tumor cells, directly enhancing their growth, but also horizontally to fibroblasts, endothelial cells and immune cells in the TME, indirectly enhancing their pro-tumor functions. TEX-driven cross-talk of MSCs with immune cells blocks their anti-tumor activity and/or converts them into suppressor cells. MSCs re-programmed by TEX mediate pro-angiogenic activity and convert stromal cells into cancer-associated fibroblasts (CAFs). Although MSCs have a potential to exert anti-tumor activities, they largely provide service to the tumor using the multidirectional communication system established by exosomes in the TME. Future therapeutic options consider disruption of this complex vicious cycle by either molecular or gene-regulated silencing of pro-tumor effects mediated by MSCs in the TME.


Assuntos
Exossomos/metabolismo , Células-Tronco Mesenquimais/imunologia , Neoplasias/imunologia , Animais , Comunicação Celular , Diferenciação Celular , Reprogramação Celular , Humanos , MicroRNAs/metabolismo , Microambiente Tumoral
19.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055057

RESUMO

Exosomes derived from tumor cells contain various molecular components, such as proteins, RNA, DNA, lipids, and carbohydrates. These components play a crucial role in all stages of tumorigenesis and development. Moreover, they reflect the physiological and pathological status of parental tumor cells. Recently, tumor-derived exosomes have become popular biomarkers for non-invasive liquid biopsy and the diagnosis of numerous cancers. The interdisciplinary significance of exosomes research has also attracted growing enthusiasm. However, the intrinsic nature of tumor-derived exosomes requires advanced methods to detect and evaluate the complex biofluid. This review analyzes the relationship between exosomes and tumors. It also summarizes the exosomal biological origin, composition, and application of molecular markers in clinical cancer diagnosis. Remarkably, this paper constitutes a comprehensive summary of the innovative research on numerous detection strategies for tumor-derived exosomes with the intent of providing a theoretical basis and reference for early diagnosis and clinical treatment of cancer.


Assuntos
Biomarcadores Tumorais , Exossomos/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Ácidos Nucleicos Livres , DNA Tumoral Circulante , Suscetibilidade a Doenças , Detecção Precoce de Câncer/métodos , Humanos , Biópsia Líquida/métodos , Técnicas de Diagnóstico Molecular , Neoplasias/etiologia , Proteínas Oncogênicas , Técnica de Seleção de Aptâmeros , Análise Espectral Raman/métodos
20.
Int J Mol Sci ; 23(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35163380

RESUMO

Exosomes are a class of small membrane-bound extracellular vesicles released by almost all cell types and present in all body fluids. Based on the studies of exosome content and their interactions with recipient cells, exosomes are now thought to mediate "targeted" information transfer. Tumor-derived exosomes (TEX) carry a cargo of molecules different from that of normal cell-derived exosomes. TEX functions to mediate distinct biological effects such as receptor discharge and intercellular cross-talk. The immune system defenses, which may initially restrict tumor progression, are progressively blunted by the broad array of TEX molecules that activate suppressive pathways in different immune cells. Herein, we provide a review of the latest research progress on TEX in the context of tumor-mediated immune suppression and discuss the potential as well as challenges of TEX as a target of immunotherapy.


Assuntos
Exossomos/metabolismo , Terapia de Imunossupressão , Neoplasias/metabolismo , Animais , Humanos , Ligantes , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA