Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cancer Sci ; 2024 Sep 22.
Article in English | MEDLINE | ID: mdl-39307979

ABSTRACT

The lack of circulating biomarkers for tumor monitoring is a major problem in Ewing sarcoma management. The development of methods for accurate tumor monitoring is required, considering the high recurrence rate of drug-resistant Ewing sarcoma. Here, we describe a sensitive analytical technique for tumor monitoring of Ewing sarcoma by detecting circulating extracellular vesicles secreted from Ewing sarcoma cells. Proteomic analysis of Ewing sarcoma cell-derived extracellular vesicles identified 564 proteins prominently observed in extracellular vesicles from three Ewing sarcoma cell lines. Among these, CD99, SLC1A5, and ENO-1 were identified on extracellular vesicles purified from sera of patients with Ewing sarcoma before treatment but not on extracellular vesicles from those after treatment and healthy individuals. Notably, not only Ewing sarcoma-derived extracellular vesicles but also Ewing sarcoma cells demonstrated proteomic expression of CD99 and ENO-1 on their surface membranes. ENO-1+CD63+ extracellular vesicle detection was reduced after tumor resection while both CD99+CD63+ and ENO-1+CD63+ extracellular vesicles were detected in serum from Ewing sarcoma-bearing mice. Finally, the accuracy of liquid biopsy targeting these candidates was assessed using extracellular vesicles from the sera of patients with Ewing sarcoma. Elevated ENO-1+CD81+ extracellular vesicles in the serum of patients before treatments distinguished patients with Ewing sarcoma from healthy individuals with an area under the curve value of 0.92 (P < 0.001) and reflected the tumor burden in patients with Ewing sarcoma during multidisciplinary treatments. Collectively, circulating ENO-1+CD81+ extracellular vesicle detection could represent a novel tool for tumor monitoring of Ewing sarcoma.

2.
Pharm Res ; 41(8): 1573-1594, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39112776

ABSTRACT

Extracellular vesicles (EVs) serve as an intrinsic system for delivering functional molecules within our body, playing significant roles in diverse physiological phenomena and diseases. Both native and engineered EVs are currently the subject of extensive research as promising therapeutics and drug delivery systems, primarily due to their remarkable attributes, such as targeting capabilities, biocompatibility, and low immunogenicity and mutagenicity. Nevertheless, their clinical application is still a long way off owing to multiple limitations. In this context, the Science Board of the Pharmaceuticals and Medical Devices Agency (PMDA) of Japan has conducted a comprehensive assessment to identify the current issues related to the quality and safety of EV-based therapeutic products. Furthermore, we have presented several examples of the state-of-the-art methodologies employed in EV manufacturing, along with guidelines for critical processes, such as production, purification, characterization, quality evaluation and control, safety assessment, and clinical development and evaluation of EV-based therapeutics. These endeavors aim to facilitate the clinical application of EVs and pave the way for their transformative impact in healthcare.


Subject(s)
Extracellular Vesicles , Quality Control , Humans , Animals , Drug Delivery Systems/methods
3.
Pharm Res ; 40(4): 889-908, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36577860

ABSTRACT

Cardiac diseases such as myocardial infarction and heart failure have been the leading cause of death worldwide for more than 20 years, and new treatments continue to be investigated. Heart transplantation, a curative treatment for severe cardiac dysfunction, is available to only a small number of patients due to the rarity of donors and high costs. Cardiac regenerative medicine using embryonic stem cells and induced pluripotent stem cells is expected to be a new alternative to heart transplantation, but it has problems such as induction of immune response, tumor formation, and low survival rate of transplanted cells. On the other hand, there has been a focus on cell-free therapy using extracellular vesicles (EVs) due to their high biocompatibility and target specificity. Exosomes, one type of EV, play a role in the molecular transport system in vivo and can be considered a drug delivery system (DDS) innate to all living things. Exosomes contain nucleic acids and proteins, which are transported from secretory cells to recipient cells. Molecules in exosomes are encapsulated in a lipid bilayer, which allows them to exist stably in body fluids without being affected by nuclease degradation enzymes. Therefore, the therapeutic use of exosomes as DDSs has been widely explored and is being used in clinical trials and other clinical settings. This review summarizes the current topics of EVs as DDSs in cardiac disease.


Subject(s)
Exosomes , Extracellular Vesicles , Heart Diseases , Neoplasms , Humans , Extracellular Vesicles/metabolism , Drug Delivery Systems , Exosomes/metabolism , Neoplasms/drug therapy
4.
J Bone Miner Metab ; 41(3): 345-357, 2023 May.
Article in English | MEDLINE | ID: mdl-35943593

ABSTRACT

Extracellular vesicles (EVs) are small particles with lipid bilayer membranes that are secreted by all cell types and are widely known as crucial intercellular communication mediators, shuttling biologically active molecules. The bone is a typically preferred site of cancer metastasis due to its unique cellular compositions and dynamics. Bone cell-derived EVs serve as regulators that orchestrate harmonious bone homeostasis. Cancer cells secrete specific EVs in a series of the bone metastatic process to dominate the bone microenvironment. Additionally, cancer cell-related EVs contribute to pre-metastatic niche formation, bone homeostasis disruption, and tumor bone progression and survival. Here, we investigated recent studies on EV-mediated crosstalk in the bone tumor microenvironment. Furthermore, this review aimed to elucidate the EV-based therapeutic perspectives for bone metastasis.


Subject(s)
Extracellular Vesicles , Extracellular Vesicles/metabolism , Cell Communication/physiology , Bone and Bones , Homeostasis
5.
Int J Mol Sci ; 24(2)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36674900

ABSTRACT

Extracellular vesicles (EVs) are particles with a lipid bilayer structure, and they are secreted by various cells in the body. EVs interact with and modulate the biological functions of recipient cells by transporting their cargoes, such as nucleic acids and proteins. EVs influence various biological phenomena, including disease progression. They also participate in tumor progression by stimulating a variety of signaling pathways and regulating immune system activation. EVs induce immune tolerance by suppressing CD8+ T-cell activation or polarizing macrophages toward the M2 phenotype, which results in tumor cell proliferation, migration, invasion, and metastasis. Moreover, immune checkpoint molecules are also expressed on the surface of EVs that are secreted by tumors that express these molecules, allowing tumor cells to not only evade immune cell attack but also acquire resistance to immune checkpoint inhibitors. During tumor metastasis, EVs contribute to microenvironmental changes in distant organs before metastatic lesions appear; thus, EVs establish a premetastatic niche. In particular, lymph nodes are adjacent organs that are connected to tumor lesions via lymph vessels, so that tumor cells metastasize to draining lymph nodes at first, such as sentinel lymph nodes. When EVs influence the microenvironment of lymph nodes, which are secondary lymphoid tissues, the immune response against tumor cells is weakened; subsequently, tumor cells spread throughout the body. In this review, we will discuss the association between EVs and tumor progression via the immune system as well as the clinical application of EVs as biomarkers and therapeutic agents.


Subject(s)
Extracellular Vesicles , Lymphatic Vessels , Humans , Lymphatic Metastasis/pathology , Extracellular Vesicles/metabolism , Cell Communication , Signal Transduction/physiology , Tumor Microenvironment/physiology
6.
Cancer Sci ; 113(10): 3498-3509, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35848896

ABSTRACT

Early detection of pancreatic ductal adenocarcinoma (PDAC) is essential for improving patient survival rates, and noninvasive biomarkers are urgently required to identify patients who are eligible for curative surgery. Here, we examined extracellular vesicles (EVs) from the serum of PDAC patients to determine their ability to detect early-stage disease. EV-associated proteins purified by ultracentrifugation and affinity columns underwent proteomic analysis to identify novel PDAC markers G protein-coupled receptor class C group 5 member C (GPRC5C) and epidermal growth factor receptor pathway substrate 8 (EPS8). To verify the potency of GPRC5C- or EPS8-positive EVs as PDAC biomarkers, we analyzed EVs from PDAC patient blood samples using ultracentrifugation in two different cohorts (a total of 54 PDAC patients, 32 healthy donors, and 22 pancreatitis patients) by immunoblotting. The combination of EV-associated GPRC5C and EPS8 had high accuracy, with area under the curve values of 0.922 and 0.946 for distinguishing early-stage PDAC patients from healthy controls in the two cohorts, respectively, and could detect PDAC patients who were negative for CA19-9. Moreover, we analyzed 30 samples taken at three time points from 10 PDAC patients who underwent surgery: before surgery, after surgery, and recurrence as an early-stage model. These proteins were detected in EVs derived from preoperative and recurrence samples. These results indicated that GPRC5C- or EPS8-positive EVs were biomarkers that have the potential to detect stage I early pancreatic cancer and small recurrent tumors detected by computed tomography.


Subject(s)
Carcinoma, Pancreatic Ductal , Extracellular Vesicles , Pancreatic Neoplasms , Adaptor Proteins, Signal Transducing , Biomarkers, Tumor , CA-19-9 Antigen , Carcinoma, Pancreatic Ductal/pathology , ErbB Receptors , Extracellular Vesicles/pathology , Humans , Pancreatic Neoplasms/pathology , Proteomics , Pancreatic Neoplasms
7.
Anal Chem ; 94(32): 11209-11215, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35797226

ABSTRACT

Extracellular vesicles (EVs) are essential intercellular communication tools, but the regulatory mechanisms governing heterogeneous EV secretion are still unclear due to the lack of methods for precise analysis. Monitoring the dynamics of secretion from individually isolated cells is crucial because in bulk analysis, secretion activity can be perturbed by cell-cell interactions, and a cell population rarely performs secretion in a magnitude- or duration-synchronized manner. Although various microfluidic techniques have been adopted to evaluate the abundance of single-cell-derived EVs, none can track their secretion dynamics continually for extended periods. Here, we have developed a droplet array-based method that allowed us to optically quantify the EV secretion dynamics of >300 single cells every 2 h for 36 h, which covers the cell doubling time of many cell types. The experimental results clearly show the highly heterogeneous nature of single-cell EV secretion and suggest that cell division facilitates EV secretion, showing the usefulness of this platform for discovering EV regulation machinery.


Subject(s)
Extracellular Vesicles , Cell Communication , Extracellular Vesicles/metabolism
8.
Cancer Cell Int ; 22(1): 367, 2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36424598

ABSTRACT

Intercellular communication plays an important role in cancer initiation and progression through direct contact and indirect interactions, such as via secretory molecules. Cancer-associated fibroblasts (CAFs) are one of the principal components of such communication with cancer cells, modulating cancer metastasis and tumour mechanics and influencing angiogenesis, the immune system, and therapeutic resistance. Over the past few years, there has been a significant increase in research on extracellular vesicles (EVs) as regulatory agents in intercellular communication. EVs enable the transfer of functional molecules, including proteins, mRNAs and microRNAs (miRNAs), to recipient cells. Cancer cells utilize EVs to dictate the specific characteristics of CAFs within the tumour microenvironment, thereby promoting cancer progression. In response to such "education" by cancer cells, CAFs contribute to cancer progression via EVs. In this review, we summarize experimental data indicating the pivotal roles of EVs in intercellular communication between cancer cells and CAFs.

9.
Am J Respir Cell Mol Biol ; 63(5): 623-636, 2020 11.
Article in English | MEDLINE | ID: mdl-32730709

ABSTRACT

Aberrant epithelial-mesenchymal interactions have critical roles in regulating fibrosis development. The involvement of extracellular vesicles (EVs), including exosomes, remains to be elucidated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). Here, we found that lung fibroblasts (LFs) from patients with IPF induce cellular senescence via EV-mediated transfer of pathogenic cargo to lung epithelial cells. Mechanistically, IPF LF-derived EVs increased mitochondrial reactive oxygen species and associated mitochondrial damage in lung epithelial cells, leading to activation of the DNA damage response and subsequent epithelial-cell senescence. We showed that IPF LF-derived EVs contain elevated levels of microRNA-23b-3p (miR-23b-3p) and miR-494-3p, which suppress SIRT3, resulting in the epithelial EV-induced phenotypic changes. Furthermore, the levels of miR-23b-3p and miR-494-3p found in IPF LF-derived EVs correlated positively with IPF disease severity. These findings reveal that the accelerated epithelial-cell mitochondrial damage and senescence observed during IPF pathogenesis are caused by a novel paracrine effect of IPF fibroblasts via microRNA-containing EVs.


Subject(s)
Cellular Senescence , Epithelial Cells/pathology , Extracellular Vesicles/metabolism , Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Aged , DNA Damage , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Lung/pathology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Models, Biological , Reactive Oxygen Species/metabolism , Sirtuin 3/metabolism
10.
Int J Mol Sci ; 21(12)2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32585976

ABSTRACT

The vast majority of cancer-related deaths are due to metastasis of the primary tumor that develops years to decades after apparent cures. However, it is difficult to effectively prevent or treat cancer metastasis. Recent studies have shown that communication between cancer cells and surrounding cells enables cancer progression and metastasis. The comprehensive term "extracellular vesicles" (EVs) describes lipid bilayer vesicles that are secreted to outside cells; EVs are well-established mediators of cell-to-cell communication. EVs participate in cancer progression and metastasis by transferring bioactive molecules, such as proteins and RNAs, including microRNAs (miRNAs), between cancer and various cells in local and distant microenvironments. Clinically, EVs functioning as diagnostic biomarkers, therapeutic targets, or even as anticancer drug-delivery vehicles have been emphasized as a result of their unique biological and pathophysiological characteristics. The potential therapeutic effects of EVs in cancer treatment are rapidly emerging and represent a new and important area of research. This review focuses on the therapeutic potential of EVs and discusses their utility for the inhibition of cancer progression, including metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Communication , Extracellular Vesicles/physiology , Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Extracellular Vesicles/drug effects , Humans , Neoplasm Metastasis , Neoplasms/pathology
11.
Int J Mol Sci ; 21(18)2020 Sep 12.
Article in English | MEDLINE | ID: mdl-32932657

ABSTRACT

Extracellular vesicles (EVs) are lipid membranous vesicles that are released from every type of cell. It has become clear that EVs are involved in a variety of biological phenomena, including cancer progression, and play critical roles in intracellular communication through the horizontal transfer of cellular cargoes such as proteins, DNA fragments, RNAs including mRNA and non-coding RNAs (microRNA, piRNA, and long non-coding RNA) and lipids. The most common cause of death associated with cancer is metastasis. Recent investigations have revealed that EVs are deeply associated with metastasis. Bone is a preferred site of metastasis, and bone metastasis is generally incurable and dramatically affects patient quality of life. Bone metastasis can cause devastating complications, including hypercalcemia, pathological fractures, spinal compression, and bone pain, which result in a poor prognosis. Although the mechanisms underlying bone metastasis have yet to be fully elucidated, increasing evidence suggests that EVs in the bone microenvironment significantly contribute to cancer progression and cancer bone tropism. Emerging evidence on EV functions in bone metastasis will facilitate the discovery of novel treatments. In this review, we will discuss the remarkable effects of EVs, especially on the tumor microenvironment in bone.


Subject(s)
Bone Neoplasms/pathology , Bone and Bones/pathology , Extracellular Vesicles/pathology , Neoplasm Metastasis/pathology , Tumor Microenvironment/physiology , Animals , Drug Delivery Systems/methods , Humans
12.
Semin Cell Dev Biol ; 67: 39-47, 2017 07.
Article in English | MEDLINE | ID: mdl-28267596

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide. Despite significant advances in lung cancer research and novel therapies, a better understanding of the disease is crucially needed to facilitate early detection and appropriate diagnoses and to improve treatment outcomes. Extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, are released from all tested cell types and modulate cell-cell communication. EVs transfer a wide variety of molecules, such as proteins, messenger RNAs and microRNAs. Emerging data suggest that EVs play an important role in lung cancer pathogenesis and may have potential as biomarkers and therapeutics. Here, we review current research on EVs in lung cancer.


Subject(s)
Extracellular Vesicles/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/metabolism , RNA, Neoplasm/genetics , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Communication , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Extracellular Vesicles/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , RNA, Neoplasm/metabolism , Signal Transduction , Translational Research, Biomedical/methods , Tumor Microenvironment/genetics
13.
Int J Mol Sci ; 20(10)2019 May 26.
Article in English | MEDLINE | ID: mdl-31130715

ABSTRACT

The primary cause of mortality among patients with cancer is the progression of the tumor, better known as cancer invasion and metastasis. Cancer progression involves a series of biologically important steps in which the cross-talk between cancer cells and the cells in the surrounding environment is positioned as an important issue. Notably, angiogenesis is a key tumorigenic phenomenon for cancer progression. Cancer-related extracellular vesicles (EVs) commonly contribute to the modulation of a microenvironment favorable to cancer cells through their function of cell-to-cell communication. Vascular-related cells such as endothelial cells (ECs) and platelets activated by cancer cells and cancer-derived EVs develop procoagulant and proinflammatory statuses, which help excite the tumor environment, and play major roles in tumor progression, including in tumor extravasation, tumor cell microthrombi formation, platelet aggregation, and metastasis. In particular, cancer-derived EVs influence ECs, which then play multiple roles such as contributing to tumor angiogenesis, loss of endothelial vascular barrier by binding to ECs, and the subsequent endothelial-to-mesenchymal transition, i.e., extracellular matrix remodeling. Thus, cell-to-cell communication between cancer cells and ECs via EVs may be an important target for controlling cancer progression. This review describes the current knowledge regarding the involvement of EVs, especially exosomes derived from cancer cells, in EC-related cancer progression.


Subject(s)
Endothelial Cells/pathology , Extracellular Vesicles/pathology , Neoplasms/pathology , Animals , Disease Progression , Exosomes/pathology , Humans , Neoplasm Metastasis/pathology , Neoplasms/blood supply , Neovascularization, Pathologic/pathology
14.
Biochem Biophys Res Commun ; 495(1): 1322-1327, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29191657

ABSTRACT

The development of drug resistance in tumor cells leads to relapse and distant metastasis. Secreted microRNAs (miRNAs) enclosed in extracellular vesicles (EVs) can act as intercellular messengers. The objective of our study was to elucidate the role of secreted miRNAs to better understand the regulatory network underlying pazopanib-resistance in synovial sarcoma cells. We performed a comprehensive analysis of secreted miRNA abundance in pazopanib treated/untreated synovial sarcoma cells from four different cell lines (SYO-1, HS-SYII, 1273/99, and YaFuSS) using microarray technology, and discovered miR-761 in EVs as a potential biomarker of pazopanib-resistance in synovial sarcoma. Furthermore, we showed that miR-761 putatively targeted three proteins, thyroid hormone receptor interactor 6 (TRIP6), lamin A/C (LMNA), and NAD-dependent protein deacetylase sirtuin-3 (SIRT3). Knockdown of any of these proteins was shown in previous studies to confer increased resistance to chemotherapeutic agents. Our findings provide new insight into the potential role of miR-761, an EV-secreted miRNA from synovial sarcoma cells, making it a potential candidate for use in sarcoma therapy in the future.


Subject(s)
Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , Pyrimidines/administration & dosage , Sarcoma, Synovial/drug therapy , Sarcoma, Synovial/metabolism , Sulfonamides/administration & dosage , Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Humans , Indazoles , LIM Domain Proteins/metabolism , Lamin Type A/metabolism , Sarcoma, Synovial/pathology , Sirtuin 3/metabolism , Transcription Factors/metabolism
15.
Biochem Biophys Res Commun ; 506(3): 723-730, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30454707

ABSTRACT

Secreted proteins enclosed in extracellular vesicles can act as intercellular messengers. The objective of this study was to elucidate the role of proteins secreted from synovial sarcoma cells in the regulatory network underlying pazopanib response. We performed a comprehensive analysis of expression of proteins secreted from four synovial sarcoma cell lines (SYO-1, HS-SYII, 1273/99, and YaFuSS) using mass spectroscopy. Comparison of up-regulated proteins in cells, extracellular vesicles-free conditioned media, and extracellular vesicles revealed significantly up-regulated Wnt in synovial sarcoma vesicles. Furthermore, we compared protein signatures of cells, conditioned media, and extracellular vesicles before and after pazopanib treatment. Interestingly, protein signatures of extracellular vesicles showed robust changes in Wnt signaling pathways in response to pazopanib. Our findings provide insight into the potential role of Wnt, a protein secreted from the extracellular vesicles of synovial sarcoma cells, making it a potential candidate for use in sarcoma diagnosis.


Subject(s)
Extracellular Vesicles/metabolism , Neoplasm Proteins/metabolism , Pyrimidines/pharmacology , Sarcoma, Synovial/metabolism , Sulfonamides/pharmacology , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Extracellular Vesicles/drug effects , Humans , Indazoles , Proteomics , Signal Transduction/drug effects
16.
Stem Cells ; 35(4): 920-931, 2017 04.
Article in English | MEDLINE | ID: mdl-27925343

ABSTRACT

Small hepatocyte-like progenitor cells (SHPCs) transiently form clusters in rat livers treated with retrorsine (Ret)/70% partial hepatectomy (PH). When Thy1+ cells isolated from d-galactosamine-treated rat livers were transplanted into the livers of Ret/PH-treated rats, the mass of the recipient liver transiently increased during the first 30 days after transplantation, suggesting that liver regeneration was enhanced. Here we addressed how Thy1+ cell transplantation stimulates liver regeneration. We found that the number and size of SHPC clusters increased in the liver at 14 days after transplantation. GeneChip analysis revealed that interleukin 17 receptor b (IL17rb) expression significantly increased in SHPCs from livers transplanted with Thy1+ cells. We subsequently searched for ligand-expressing cells and found that sinusoidal endothelial cells (SECs) and Kupffer cells expressed Il17b and Il25, respectively. Moreover, extracellular vesicles (EVs) separated from the conditioned medium of Thy1+ cell culture induced IL17b and IL25 expression in SECs and Kupffer cells, respectively. Furthermore, EVs enhanced IL17rb expression in small hepatocytes (SHs), which are hepatocytic progenitor cells; in culture, IL17B stimulated the growth of SHs. These results suggest that Thy1-EVs coordinate IL17RB signaling to enhance liver regeneration by targeting SECs, Kupffer cells, and SHPCs. Indeed, the administration of Thy1-EVs increased the number and size of SHPC clusters in Ret/PH-treated rat livers. Sixty days post-transplantation, most expanded SHPCs entered cellular senescence, and the enlarged liver returned to its normal size. In conclusion, Thy1+ cell transplantation enhanced liver regeneration by promoting the proliferation of intrinsic hepatic progenitor cells via IL17RB signaling. Stem Cells 2017;35:920-931.


Subject(s)
Cell Transplantation , Hepatocytes/cytology , Hepatocytes/metabolism , Liver Regeneration , Receptors, Interleukin-17/metabolism , Signal Transduction , Stem Cells/cytology , Thy-1 Antigens/metabolism , Animals , Cell Aggregation/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Cellular Senescence/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Vesicles/drug effects , Extracellular Vesicles/metabolism , Gene Expression Regulation/drug effects , Hepatectomy , Hepatocytes/drug effects , Interleukin-17/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/cytology , Liver Regeneration/drug effects , Male , Pyrrolizidine Alkaloids/pharmacology , Rats, Inbred F344 , Signal Transduction/drug effects , Stem Cells/drug effects , Stem Cells/metabolism
17.
Jpn J Clin Oncol ; 48(10): 869-876, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30169666

ABSTRACT

Over the past several years, considerable focus has been placed on the need for 'biomarkers'. However, traditional biomarkers, such as CEA or CA19-9, for gastrointestinal cancer do not provide sufficient sensitivity and specificity for diagnosing cancer. Moreover, these biomarkers cannot provide information regarding the individual variability of patients. Recently, extracellular vesicles (EVs) and extracellular microRNA (miRNA) have shown potential in cancer diagnosis because tumor cells have been shown to release EVs and miRNAs, which mirror their cellular origin, into circulation. Therefore, detection of tumor-specific EVs and extracellular miRNA in body fluids from cancer patients could serve as a non-invasive liquid biopsy for cancer diagnosis and monitoring. This review explores the potential contribution of liquid biopsy using EVs and extracellular miRNA to diagnosis and monitor cancer, including an assessment of prognosis and early detection of disease recurrence in patients with cancer.


Subject(s)
Biomarkers, Tumor/genetics , Extracellular Vesicles/metabolism , Liquid Biopsy/methods , MicroRNAs/genetics , Humans , Prognosis
18.
Mol Ther ; 25(1): 181-191, 2017 01 04.
Article in English | MEDLINE | ID: mdl-28129113

ABSTRACT

Metastasis is the main cause of cancer mortality for many types of cancer; however, difficulties remain in effectively preventing metastasis. It has been recently and widely reported that cancer-derived extracellular vesicles (EVs) contribute to cancer metastasis. Thus, therapeutic strategies targeting cancer-derived EVs hold great promise because of the possibility of EVs driving the cancer microenvironment toward metastasis. Here, we provide a novel strategy for therapeutic antibody treatment to target cancer-derived EVs and inhibit the metastasis of breast cancer in a mouse model, establishing a rationale for further clinical investigation. Treatment with human-specific anti-CD9 or anti-CD63 antibodies significantly decreased metastasis to the lungs, lymph nodes, and thoracic cavity, although no obvious effects on primary xenograft tumor growths were observed. In in vitro and in vivo experiments, the EVs incubated with the targeted antibodies were preferentially internalized by macrophages, suggesting that antibody-tagged cancer-derived EVs would be eliminated by macrophages. Our results suggested that therapeutic antibody administration effectively suppresses EV-triggered metastasis in cancer and that the removal of EVs could be a novel strategy for cancer therapy.


Subject(s)
Antineoplastic Agents , Extracellular Vesicles/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Antibodies, Monoclonal/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell-Derived Microparticles/metabolism , Disease Models, Animal , Extracellular Vesicles/immunology , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/therapy , Phagocytosis , Tetraspanin 29/immunology , Tetraspanin 29/metabolism , Tetraspanin 30/immunology , Tetraspanin 30/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology , Xenograft Model Antitumor Assays
19.
Cell Mol Life Sci ; 74(4): 697-713, 2017 02.
Article in English | MEDLINE | ID: mdl-27582126

ABSTRACT

Intercellular communication plays an important role in cancer initiation and progression through secretory molecules, including growth factors and cytokines. Recent advances have revealed that small membrane vesicles, termed extracellular vesicles (EVs), served as a regulatory agent in the intercellular communication of cancer. EVs enable the transfer of functional molecules, including proteins, mRNA and microRNAs (miRNAs), into recipient cells. Cancer cells utilize EVs to dictate the unique phenotype of surrounding cells, thereby promoting cancer progression. Against such "education" by cancer cells, non-tumoral cells suppress cancer initiation and progression via EVs. Therefore, researchers consider EVs to be important cues to clarify the molecular mechanisms of cancer biology. Understanding the functions of EVs in cancer progression is an important aspect of cancer biology that has not been previously elucidated. In this review, we summarize experimental data that indicate the pivotal roles of EVs in cancer progression.


Subject(s)
Cell Communication , Exosomes/pathology , Extracellular Vesicles/pathology , Neoplasms/pathology , Tumor Microenvironment , Animals , Disease Progression , Exosomes/immunology , Exosomes/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/metabolism , Humans , Immune System/immunology , Immune System/metabolism , Immune System/pathology , Immunity, Cellular , Neoplasms/immunology , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL