Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.058
Filter
1.
Neurol India ; 72(2): 285-291, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38691471

ABSTRACT

BACKGROUND: Microparticles (MPs) have been implicated in thrombosis and endothelial dysfunction. Their involvement in early coagulopathy and in worsening of outcomes in isolated severe traumatic brain injury (sTBI) patients remains ill defined. OBJECTIVE: We sought to quantify the circulatory MP subtypes derived from platelets (PMPs; CD42), endothelial cells (EMPs; CD62E), and those bearing tissue factor (TFMP; CD142) and analyze their correlation with early coagulopathy, thrombin generation, and in-hospital mortality. MATERIALS AND METHODS: Prospective screening of sTBI patients was done. Blood samples were collected before blood and fluid transfusion. MP enumeration and characterization were performed using flow cytometry, and thrombin-antithrombin complex (TAT) levels were determined using enzyme-linked immunosorbent assay (ELISA). Circulating levels of procoagulant MPs were compared between isolated sTBI patients and age- and gender-matched healthy controls (HC). Patients were stratified according to their PMP, EMP, and TFMP levels, respectively (high ≥HC median and low < HC median). RESULTS: Isolated sTBI resulted in an increased generation of PMPs (456.6 [228-919] vs. 249.1 [198.9-404.5]; P = 0.01) and EMPs (301.5 [118.8-586.7] vs. 140.9 [124.9-286]; P = 0.09) compared to HCs. Also, 5.3% of MPs expressed TF (380 [301-710]) in HCs, compared to 6.6% MPs (484 [159-484]; P = 0.87) in isolated sTBI patients. Early TBI-associated coagulopathy (TBI-AC) was seen in 50 (41.6%) patients. PMP (380 [139-779] vs. 523.9 [334-927]; P = 0.19) and EMP (242 [86-483] vs. 344 [168-605]; P = 0.81) counts were low in patients with TBI-AC, compared to patients without TBI-AC. CONCLUSION: Our results suggest that enhanced cellular activation and procoagulant MP generation are predominant after isolated sTBI. TBI-AC was associated with low plasma PMPs count compared to the count in patients without TBI-AC. Low PMPs may be involved with the development of TBI-AC.


Subject(s)
Blood Coagulation Disorders , Brain Injuries, Traumatic , Cell-Derived Microparticles , Humans , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/mortality , Cell-Derived Microparticles/metabolism , Female , Male , Adult , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/blood , Middle Aged , Prospective Studies , Thromboplastin/metabolism , Blood Platelets/metabolism , Hospital Mortality , Endothelial Cells/metabolism
2.
J Nanobiotechnology ; 22(1): 225, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38705987

ABSTRACT

Immunogenic cell death (ICD) plays a crucial role in triggering the antitumor immune response in the tumor microenvironment (TME). Recently, considerable attention has been dedicated to ferroptosis, a type of ICD that is induced by intracellular iron and has been demonstrated to change the immune desert status of the TME. However, among cancers that are characterized by an immune desert, such as prostate cancer, strategies for inducing high levels of ferroptosis remain limited. Radiated tumor cell-derived microparticles (RMPs) are radiotherapy mimetics that have been shown to activate the cGAS-STING pathway, induce tumor cell ferroptosis, and inhibit M2 macrophage polarization. RMPs can also act as carriers of agents with biocompatibility. In the present study, we designed a therapeutic system wherein the ferroptosis inducer RSL-3 was loaded into RMPs, which were tested in in vitro and in vivo prostate carcinoma models established using RM-1 cells. The apoptosis inducer CT20 peptide (CT20p) was also added to the RMPs to aggravate ferroptosis. Our results showed that RSL-3- and CT20p-loaded RMPs (RC@RMPs) led to ferroptosis and apoptosis of RM-1 cells. Moreover, CT20p had a synergistic effect on ferroptosis by promoting reactive oxygen species (ROS) production, lipid hydroperoxide production, and mitochondrial instability. RC@RMPs elevated dendritic cell (DC) expression of MHCII, CD80, and CD86 and facilitated M1 macrophage polarization. In a subcutaneously transplanted RM-1 tumor model in mice, RC@RMPs inhibited tumor growth and prolonged survival time via DC activation, macrophage reprogramming, enhancement of CD8+ T cell infiltration, and proinflammatory cytokine production in the tumor. Moreover, combination treatment with anti-PD-1 improved RM-1 tumor inhibition. This study provides a strategy for the synergistic enhancement of ferroptosis for prostate cancer immunotherapies.


Subject(s)
Cell-Derived Microparticles , Ferroptosis , Prostatic Neoplasms , Reactive Oxygen Species , Tumor Microenvironment , Ferroptosis/drug effects , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Animals , Mice , Cell-Derived Microparticles/metabolism , Cell Line, Tumor , Humans , Reactive Oxygen Species/metabolism , Macrophages/metabolism , Macrophages/drug effects , Apoptosis/drug effects , Mice, Inbred C57BL
3.
J Nanobiotechnology ; 22(1): 237, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735920

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) promote tumor growth, metastasis, and lead to immunotherapy resistance. Studies revealed that miRNAs are also expressed in MDSCs and promote the immunosuppressive function of MDSCs. Currently, few studies have been reported on inducible cellular microvesicle delivery of nucleic acid drugs targeting miRNA in MDSCs for the treatment of malignant tumors. RESULTS AND CONCLUSION: In this study, we designed an artificial DNA named G-quadruplex-enhanced circular single-stranded DNA-9 (G4-CSSD9), that specifically adsorbs the miR-9 sequence. Its advanced DNA folding structure, rich in tandem repeat guanine (G-quadruplex), also provides good stability. Mesenchymal stem cells (MSCs) were prepared into nanostructured vesicles by membrane extrusion. The MSC microvesicles-encapsulated G4-CSSD9 (MVs@G4-CSSD9) was delivered into MDSCs, which affected the downstream transcription and translation process, and reduced the immunosuppressive function of MDSCs, so as to achieve the purpose of treating melanoma. In particular, it provides an idea for the malignant tumor treatment.


Subject(s)
DNA, Single-Stranded , G-Quadruplexes , Mesenchymal Stem Cells , MicroRNAs , Myeloid-Derived Suppressor Cells , Animals , Myeloid-Derived Suppressor Cells/metabolism , Mice , DNA, Single-Stranded/chemistry , Cell Line, Tumor , Mice, Inbred C57BL , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/metabolism , DNA, Circular/chemistry , Humans , Melanoma/drug therapy
4.
Mol Biol Rep ; 51(1): 676, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796661

ABSTRACT

BACKGROUND: The current understanding emphasizes the intricate interplay between the Leukemic cell and its environment. Platelet-derived microparticles play a crucial role in facilitating intercellular communication and contribute to the complex landscape of cancer pathology. This study aimed to investigate the influence of platelet-derived microparticles on cell proliferation, apoptosis, and the expression of key genes, including P53, P21, Cyclin D1, Bax, and Bcl-2, within the context of a chronic myeloid leukemia cell line (K562). METHODS AND RESULTS: Platelet-derived microparticles were obtained through centrifugation at various speeds, and their concentration was quantified using the BCA assay. To determine the size and immunophenotypic characteristics of the PMPs, both the DLS technique and flow cytometry were employed. Cell proliferation was assessed using the MTT assay and hemocytometer, and cell cycle analysis was conducted through DNA content evaluation. Real-time PCR was utilized for gene expression analysis of Bax, Bcl-2, Cyclin D1, P53, and P21. Flow cytometry was employed to examine cell apoptosis. The findings revealed that platelet-derived microparticles have the ability to decrease proliferation of the K562 cell line, while not exerting an impact on apoptosis and cell cycle progression. Analysis through real-time PCR indicated an upregulation in the gene expression of P53, P21, and Bcl-2, accompanied by a downregulation in Bax and Cyclin D1. CONCLUSION: This investigation sheds light on the intricate relationship between chronic myeloid leukemia and its microenvironment, particularly the involvement of platelet-derived microparticles. The study underscores the potential of platelet-derived microparticles to influence cell behavior and gene expression, providing a deeper understanding of their role in CML and its therapeutic implications.


Subject(s)
Apoptosis , Blood Platelets , Cell Proliferation , Cell-Derived Microparticles , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Cell-Derived Microparticles/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Blood Platelets/metabolism , K562 Cells , Cell Proliferation/genetics , Apoptosis/genetics , Cell Cycle/genetics , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Cyclin D1/metabolism , Cyclin D1/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Gene Expression Regulation, Leukemic
5.
J Am Heart Assoc ; 13(9): e032698, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38690710

ABSTRACT

BACKGROUND: Provoked anger is associated with an increased risk of cardiovascular disease events. The underlying mechanism linking provoked anger as well as other core negative emotions including anxiety and sadness to cardiovascular disease remain unknown. The study objective was to examine the acute effects of provoked anger, and secondarily, anxiety and sadness on endothelial cell health. METHODS AND RESULTS: Apparently healthy adult participants (n=280) were randomized to an 8-minute anger recall task, a depressed mood recall task, an anxiety recall task, or an emotionally neutral condition. Pre-/post-assessments of endothelial health including endothelium-dependent vasodilation (reactive hyperemia index), circulating endothelial cell-derived microparticles (CD62E+, CD31+/CD42-, and CD31+/Annexin V+) and circulating bone marrow-derived endothelial progenitor cells (CD34+/CD133+/kinase insert domain receptor+ endothelial progenitor cells and CD34+/kinase insert domain receptor+ endothelial progenitor cells) were measured. There was a group×time interaction for the anger versus neutral condition on the change in reactive hyperemia index score from baseline to 40 minutes (P=0.007) with a mean±SD change in reactive hyperemia index score of 0.20±0.67 and 0.50±0.60 in the anger and neutral conditions, respectively. For the change in reactive hyperemia index score, the anxiety versus neutral condition group by time interaction approached but did not reach statistical significance (P=0.054), and the sadness versus neutral condition group by time interaction was not statistically significant (P=0.160). There were no consistent statistically significant group×time interactions for the anger, anxiety, and sadness versus neutral condition on endothelial cell-derived microparticles and endothelial progenitor cells from baseline to 40 minutes. CONCLUSIONS: In this randomized controlled experimental study, a brief provocation of anger adversely affected endothelial cell health by impairing endothelium-dependent vasodilation.


Subject(s)
Anger , Anxiety , Endothelium, Vascular , Vasodilation , Humans , Male , Female , Adult , Endothelium, Vascular/physiopathology , Anxiety/psychology , Endothelial Progenitor Cells/metabolism , Middle Aged , Sadness , Cell-Derived Microparticles/metabolism , Hyperemia/physiopathology , Emotions , Young Adult , Time Factors , Endothelial Cells
6.
J Nanobiotechnology ; 22(1): 201, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38659058

ABSTRACT

The utilization of extracellular vesicles (EV) in immunotherapy, aiming at suppressing peripheral immune cells responsible for inflammation, has demonstrated significant efficacy in treating various inflammatory diseases. However, the clinical application of EV has faced challenges due to their inadequate targeting ability. In addition, most of the circulating EV would be cleared by the liver, resulting in a short biological half-life after systemic administration. Inspired by the natural microvesicles (MV, as a subset of large size EV) are originated and shed from the plasma membrane, we developed the immunosuppressive MV-mimetic (MVM) from endotoxin tolerant dendritic cells (DC) by a straightforward and effective extrusion approach, in which DC surface proteins were inherited for providing the homing ability to the spleen, while αCD3 antibodies were conjugated to the MVM membranes for specific targeting of T cells. The engineered MVM carried a large number of bioactive cargos from the parental cells, which exhibited a remarkable ability to promote the induction of regulatory T cells (Treg) and polarization of anti-inflammatory M2 macrophages. Mechanistically, the elevated Treg level by MVM was mediated due to the upregulation of miR-155-3p. Furthermore, it was observed that systemic and local immunosuppression was induced by MVM in models of sepsis and rheumatoid arthritis through the improvement of Treg and M2 macrophages. These findings reveal a promising cell-free strategy for managing inflammatory responses to infections or tissue injury, thereby maintaining immune homeostasis.


Subject(s)
Cell-Derived Microparticles , Dendritic Cells , Inflammation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Mice , Inflammation/drug therapy , Cell-Derived Microparticles/metabolism , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/drug effects , Extracellular Vesicles , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Sepsis/immunology , Sepsis/drug therapy , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Humans , Immunotherapy/methods
7.
Iran J Kidney Dis ; 18(2): 99-107, 2024 03.
Article in English | MEDLINE | ID: mdl-38660698

ABSTRACT

INTRODUCTION: We recently discovered that microvesicles (MVs)  derived from mesenchymal stem cells (MSCs) overexpressing  miRNA-34a can alleviate experimental kidney injury in mice. In  this study, we further explored the effects of miR34a-MV on renal  fibrosis in the unilateral ureteral obstruction (UUO) models.  Methods. Bone marrow MSCs were modified by lentiviruses  overexpressing miR-34a, and MVs were collected from the  supernatants of MSCs. C57BL6/J mice were divided into control,  unilateral ureteral obstruction (UUO), UUO + MV, UUO + miR-34aMV and UUO + miR-34a-inhibitor-MV groups. MVs were injected  to mice after surgery. The mice were then euthanized on day 7  and 14 of modeling, and renal tissues were collected for further  analyses by Hematoxylin and eosin, Masson's trichrome,  and Immunohistochemical (IHC) staining.  Results. The UUO + MV group exhibited a significantly reduced  degree of renal interstitial fibrosis with inflammatory cell infiltration,  tubular epithelial cell atrophy, and vacuole degeneration compared  with the UUO group. Surprisingly, overexpressing miR-34a enhanced  these effects of MSC-MV on the UUO mice.  Conclusion. Our study demonstrates that miR34a further enhances  the effects of MSC-MV on renal fibrosis in mice through the  regulation of epithelial-to-mesenchymal transition (EMT) and  Notch pathway. miR-34a may be a candidate molecular therapeutic  target for the treatment of renal fibrosis. DOI: 10.52547/ijkd.7673.


Subject(s)
Cell-Derived Microparticles , Kidney Diseases , Kidney , Mesenchymal Stem Cells , MicroRNAs , Animals , Male , Mice , Cell-Derived Microparticles/genetics , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/transplantation , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Kidney/pathology , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Signal Transduction , Ureteral Obstruction
8.
Cancer Lett ; 592: 216898, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38670306

ABSTRACT

Radiotherapy (RT) is used for over 50 % of cancer patients and can promote adaptive immunity against tumour antigens. However, the underlying mechanisms remain unclear. Here, we discovered that RT induces the release of irradiated tumour cell-derived microparticles (RT-MPs), which significantly upregulate MHC-I expression on the membranes of non-irradiated cells, enhancing the recognition and killing of these cells by T cells. Mechanistically, RT-MPs induce DNA double-strand breaks (DSB) in tumour cells, activating the ATM/ATR/CHK1-mediated DNA repair signalling pathway, and upregulating MHC-I expression. Inhibition of ATM/ATR/CHK1 reversed RT-MP-induced upregulation of MHC-I. Furthermore, phosphorylation of STAT1/3 following the activation of ATM/ATR/CHK1 is indispensable for the DSB-dependent upregulation of MHC-I. Therefore, our findings reveal the role of RT-MP-induced DSBs and the subsequent DNA repair signalling pathway in MHC-I expression and provide mechanistic insights into the regulation of MHC-I expression after DSBs.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Cell-Derived Microparticles , DNA Breaks, Double-Stranded , DNA Repair , Histocompatibility Antigens Class I , Signal Transduction , Up-Regulation , Humans , Cell-Derived Microparticles/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 1/genetics , Animals , Phosphorylation , Gene Expression Regulation, Neoplastic , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Mice , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/radiotherapy , Neoplasms/immunology
9.
Br J Haematol ; 204(5): 2025-2039, 2024 May.
Article in English | MEDLINE | ID: mdl-38613149

ABSTRACT

Splenectomised ß-thalassaemia/haemoglobin E (HbE) patients have increased levels of circulating microparticles or medium extra-cellular vesicles (mEVs). The splenectomised mEVs play important roles in thromboembolic complications in patients since they can induce platelet activation and endothelial cell dysfunction. However, a comprehensive understanding of the mechanism of mEV generation in thalassaemia disease has still not been reached. Thalassaemic mEVs are hypothesised to be generated from cellular oxidative stress in red blood cells (RBCs) and platelets. Therefore, a proteomic analysis of mEVs from splenectomised and non-splenectomised ß-thalassaemia/HbE patients was performed by liquid chromatography with tandem mass spectrometry. A total of 171 proteins were identified among mEVs. Interestingly, 72 proteins were uniquely found in splenectomised mEVs including immunoglobulin subunits and cytoskeleton proteins. Immunoglobulin G (IgG)-bearing mEVs in splenectomised patients were significantly increased. Furthermore, complement C1q was detected in both mEVs with IgG binding and mEVs without IgG binding. Interestingly, the percentage of mEVs generated from RBCs with IgG binding was approximately 15-20 times higher than the percentage of RBCs binding with IgG. This suggested that the vesiculation of thalassaemia mEVs could be a mechanism of RBCs to eliminate membrane patches harbouring immune complex and may consequently prevent cells from phagocytosis and lysis.


Subject(s)
Hemoglobin E , Proteomics , beta-Thalassemia , Humans , beta-Thalassemia/blood , beta-Thalassemia/metabolism , Hemoglobin E/metabolism , Proteomics/methods , Female , Male , Adult , Extracellular Vesicles/metabolism , Splenectomy , Immunoglobulin G/blood , Erythrocyte Membrane/metabolism , Proteome/analysis , Adolescent , Erythrocytes/metabolism , Cell-Derived Microparticles/metabolism , Young Adult
11.
Proteomics ; 24(11): e2300058, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38470197

ABSTRACT

Previously, we reported that human primary (SW480) and metastatic (SW620) colorectal (CRC) cells release three classes of membrane-encapsulated extracellular vesicles (EVs); midbody remnants (MBRs), exosomes (Exos), and microparticles (MPs). We reported that MBRs were molecularly distinct at the protein level. To gain further biochemical insights into MBRs, Exos, and MPs and their emerging role in CRC, we performed, and report here, for the first time, a comprehensive transcriptome and long noncoding RNA sequencing analysis and fusion gene identification of these three EV classes using the next-generation RNA sequencing technique. Differential transcript expression analysis revealed that MBRs have a distinct transcriptomic profile compared to Exos and MPs with a high enrichment of mitochondrial transcripts lncRNA/pseudogene transcripts that are predicted to bind to ribonucleoprotein complexes, spliceosome, and RNA/stress granule proteins. A salient finding from this study is a high enrichment of several fusion genes in MBRs compared to Exos, MPs, and cell lysates from their parental cells such as MSH2 (gene encoded DNA mismatch repair protein MSH2). This suggests potential EV-liquid biopsy targets for cancer detection. Importantly, the expression of cancer progression-related transcripts found in EV classes derived from SW480 (EGFR) and SW620 (MET and MACCA1) cell lines reflects their parental cell types. Our study is the report of RNA and fusion gene compositions within MBRs (including Exos and MPs) that could have an impact on EV functionality in cancer progression and detection using EV-based RNA/ fusion gene candidates for cancer biomarkers.


Subject(s)
Colorectal Neoplasms , Exosomes , Gene Expression Profiling , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Exosomes/genetics , Exosomes/metabolism , Gene Expression Profiling/methods , Cell Line, Tumor , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/genetics , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Transcriptome/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
12.
Mar Drugs ; 22(2)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38393026

ABSTRACT

Chondrosia reniformis is a collagen-rich marine sponge that is considered a sustainable and viable option for producing an alternative to mammalian-origin collagens. However, there is a lack of knowledge regarding the properties of collagen isolated from different sponge parts, namely the outer region, or cortex, (ectosome) and the inner region (choanosome), and how it affects the development of biomaterials. In this study, a brief histological analysis focusing on C. reniformis collagen spatial distribution and a comprehensive comparative analysis between collagen isolated from ectosome and choanosome are presented. The isolated collagen characterization was based on isolation yield, Fourier-transformed infrared spectroscopy (FTIR), circular dichroism (CD), SDS-PAGE, dot blot, and amino acid composition, as well as their cytocompatibility envisaging the development of future biomedical applications. An isolation yield of approximately 20% was similar for both sponge parts, as well as the FTIR, CD, and SDS-PAGE profiles, which demonstrated that both isolated collagens presented a high purity degree and preserved their triple helix and fibrillar conformation. Ectosome collagen had a higher OHpro content and possessed collagen type I and IV, while the choanosome was predominately constituted by collagen type IV. In vitro cytotoxicity assays using the L929 fibroblast cell line displayed a significant cytotoxic effect of choanosome collagen at 2 mg/mL, while ectosome collagen enhanced cell metabolism and proliferation, thus indicating the latter as being more suitable for the development of biomaterials. This research represents a unique comparative study of C. reniformis body parts, serving as a support for further establishing this marine sponge as a promising alternative collagen source for the future development of biomedical applications.


Subject(s)
Cell-Derived Microparticles , Porifera , Animals , Cell-Derived Microparticles/metabolism , Biocompatible Materials/pharmacology , Biocompatible Materials/metabolism , Porifera/metabolism , Collagen/chemistry , Collagen Type I/metabolism , Mammals/metabolism
13.
Oncoimmunology ; 13(1): 2304963, 2024.
Article in English | MEDLINE | ID: mdl-38235317

ABSTRACT

Approximately two-thirds of hepatocellular carcinoma (HCC) is considered a "cold tumor" characterized by few tumor-infiltrating T cells and an abundance of immunosuppressive cells. Cilengitide, an integrin αvß3 inhibitor, has failed in clinical trials as a potential anticancer drug. This failure implies that integrin αvß3 may play an important role in immune cells. However, the expression and potential role of integrin αvß3 in T cells of HCC patients remain unknown. Here, we established two HCC models and found that cilengitide had a dual effect on the HCC microenvironment by exerting both antitumor effect and immunosuppressive effect on T cells. This may partly explain the failure of cilengitide in clinical trials. In clinical specimens, HCC-infiltrating T cells exhibited deficient expression and activation of integrin ß3, which was associated with poor T-cell infiltration into tumors. Additionally, integrin ß3 functioned as a positive immunomodulatory molecule to facilitate T-cell infiltration and T helper 1-type immune response in vitro. Furthermore, T cells and platelet-derived microparticles (PMPs) co-culture assay revealed that PMPs adoptively transferred integrin ß3 to T cells and positively regulated T cell immune response. This process was mediated by clathrin-dependent endocytosis and macropinocytosis. Our data demonstrate that integrin ß3 deficiency on HCC-infiltrating T cells may be involved in shaping the immunosuppressive tumor microenvironment. PMPs transfer integrin ß3 to T cells and positively regulate T cell immune response, which may provide a new insight into immune therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell-Derived Microparticles , Liver Neoplasms , Humans , Integrin beta3/metabolism , Integrin beta3/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Integrin alphaVbeta3/therapeutic use , Liver Neoplasms/drug therapy , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , T-Lymphocytes , Tumor Microenvironment
14.
Biomater Sci ; 12(5): 1131-1150, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38284828

ABSTRACT

Extracellular vesicles (EVs) are vesicles with lipid bilayer structures shed from the plasma membrane of cells. Microvesicles (MVs) are a subset of EVs containing proteins, lipids, nucleic acids, and other metabolites. MVs can be produced under specific cell stimulation conditions and isolated by modern separation technology. Due to their tumor homing and large volume, tumor cell-derived microvesicles (TMVs) have attracted interest recently and become excellent delivery carriers for therapeutic vaccines, imaging agents or antitumor drugs. However, preparing sufficient and high-purity TMVs and conducting clinical transformation has become a challenge in this field. In this review, the recent research achievements in the generation, isolation, characterization, modification, and application of TMVs in cancer therapy are reviewed, and the challenges facing therapeutic applications are also highlighted.


Subject(s)
Cell-Derived Microparticles , Extracellular Vesicles , Neoplasms , Humans , Cell-Derived Microparticles/chemistry , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/pathology , Extracellular Vesicles/chemistry , Neoplasms/drug therapy , Cell Membrane
15.
Mol Cancer Res ; 22(3): 268-281, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38085263

ABSTRACT

An increasing number of studies show that platelets as well as platelet-derived microparticles (PMP) play significant roles in cancer malignancy and disease progression. Particularly, PMPs have the capacity to interact and internalize within target cells resulting in the transfer of their bioactive cargo, which can modulate the signaling and activation processes of recipient cells. We recently identified a new subpopulation of these vesicles (termed mitoMPs), which contain functional mitochondria. Given the predominant role of mitochondria in cancer cell metabolism and disease progression, we set out to investigate the impact of mitoMPs on breast cancer metabolic reprograming and phenotypic processes leading to malignancy. Interestingly, we observed that recipient cell permeability to PMP internalization varied among the breast cancer cell types evaluated in our study. Specifically, cells permissive to mitoMPs acquire mitochondrial-dependent functions, which stimulate increased cellular oxygen consumption rates and intracellular ATP levels. In addition, cancer cells co-incubated with PMPs display enhanced malignant features in terms of migration and invasion. Most importantly, the cancer aggressive processes and notable metabolic plasticity induced by PMPs were highly dependent on the functional status of the mitoMP-packaged mitochondria. These findings characterize a new mechanism by which breast cancer cells acquire foreign mitochondria resulting in the gain of metabolic processes and malignant features. A better understanding of these mechanisms may provide therapeutic opportunities through PMP blockade to deprive cancer cells from resources vital in disease progression. IMPLICATIONS: We show that the transfer of foreign mitochondria by microparticles modulates recipient cancer cell metabolic plasticity, leading to greater malignant processes.


Subject(s)
Breast Neoplasms , Cell-Derived Microparticles , Humans , Female , Breast Neoplasms/metabolism , Cell-Derived Microparticles/metabolism , Energy Metabolism , Mitochondria/metabolism , Disease Progression
16.
Int J Mol Sci ; 24(21)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37958844

ABSTRACT

Mesenchymal stem cells (MSCs) and their derivatives can be promising tools in oncology including ovarian cancer treatment. This study aimed to determine the effect of HATMSC2-MVs (microvesicles derived from human immortalized mesenchymal stem cells of adipose tissue origin) on the fate and behavior of primary ovarian cancer cells. Human primary ovarian cancer (OvCa) cells were isolated from two sources: post-operative tissue of ovarian cancer and ascitic fluid. The phenotype of cells was characterized using flow cytometry, real-time RT-PCR, and immunofluorescence staining. The effect of HATMSC2-MVs on the biological activity of primary cells was analyzed in 2D (proliferation, migration, and cell survival) and 3D (cell survival) models. We demonstrated that HATMSC2-MVs internalized into primary ovarian cancer cells decrease the metabolic activity and induce the cancer cell death and are leading to decreased migratory activity of tumor cells. The results suggests that the anti-cancer effect of HATMSC2-MVs, with high probability, is contributed by the delivery of molecules that induce cell cycle arrest and apoptosis (p21, tumor suppressor p53, executor caspase 3) and proapoptotic regulators (bad, BIM, Fas, FasL, p27, TRAIL-R1, TRAIL-R2), and their presence has been confirmed by apoptotic protein antibody array. In this study, we demonstrate the ability to inhibit primary OvCa cells growth and apoptosis induction after exposure of OvCa cells on HATMSC2-MVs treatment; however, further studies are needed to clarify their anticancer activities.


Subject(s)
Cell-Derived Microparticles , Mesenchymal Stem Cells , Ovarian Neoplasms , Humans , Female , Mesenchymal Stem Cells/metabolism , Apoptosis , Adipose Tissue , Ovarian Neoplasms/metabolism , Cell-Derived Microparticles/metabolism
17.
Cell Rep Med ; 4(12): 101303, 2023 12 19.
Article in English | MEDLINE | ID: mdl-38029750

ABSTRACT

The inadequate activation of antigen-presenting cells, the entanglement of T cells, and the highly immunosuppressive conditions in the tumor microenvironment (TME) are important factors that limit the effectiveness of cancer vaccines. Studies show that a personalized and broad antigen repertoire fully activates anti-tumor immunity and that inhibiting the function of transforming growth factor (TGF)-ß facilitates T cell migration. In our study, we introduce a vaccine strategy by engineering irradiated tumor cell-derived microparticles (RT-MPs), which have both personalized and broad antigen repertoire, to induce comprehensive anti-tumor effects. Encouraged by the proinflammatory effects of the spike protein from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the high affinity between TGF-ß receptor 2 (TGFBR2) and TGF-ß, we develop RT-MPs with the SARS-CoV-2 spike protein and TGFBR2. This spike protein and high TGFBR2 expression induce the innate immune response and ameliorate the immunosuppressive TME, thereby promoting T cell activation and infiltration and ultimately inhibiting tumor growth. Our study provides a strategy for producing an effective personalized anti-tumor vaccine.


Subject(s)
Cancer Vaccines , Cell-Derived Microparticles , Neoplasms , Humans , Spike Glycoprotein, Coronavirus , Receptor, Transforming Growth Factor-beta Type II , Cell-Derived Microparticles/metabolism , Neoplasms/therapy , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
18.
Drug Discov Today ; 28(11): 103791, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37777169

ABSTRACT

Prostate cancer (PCa) is the second most common and fifth most aggressive neoplasm among men worldwide. In the last decade, extracellular vesicle (EV) research has decoded multiple unsolved cancer-related mysteries. EVs can be classified as microvesicles, apoptotic bodies, and exosomes, among others. Exosomes play a key role in cellular signaling. Their internal cargos (nucleic acids, proteins, lipids) influence the recipient cell. In PCa, the exosome is the regulator of cancer progression. It is also a promising theranostics tool for PCa. Moreover, exosomes have strong participation in male fertility complications. This review aims to highlight the exosome theranostics signature in PCa and its association with male fertility.


Subject(s)
Cell-Derived Microparticles , Exosomes , Extracellular Vesicles , Prostatic Neoplasms , Humans , Male , Extracellular Vesicles/metabolism , Cell-Derived Microparticles/metabolism , Fertility
19.
Int J Mol Sci ; 24(15)2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37569299

ABSTRACT

Aside from their key protective roles in hemostasis and innate immunity, platelets are now recognized as having multifaceted, adverse roles in the pathogenesis, progression and outcome of many types of human malignancy. The most consistent and compelling evidence in this context has been derived from the notable association of elevated circulating platelet counts with the onset and prognosis of various human malignancies, particularly lung cancer, which represents the primary focus of the current review. Key topics include an overview of the association of lung cancer with the circulating platelet count, as well as the mechanisms of platelet-mediated, pro-tumorigenic immunosuppression, particularly the role of transforming growth factor beta 1. These issues are followed by a discussion regarding the pro-tumorigenic role of platelet-derived microparticles (PMPs), the most abundant type of microparticles (MPs) in human blood. In this context, the presence of increased levels of PMPs in the blood of lung cancer patients has been associated with tumor growth, invasion, angiogenesis and metastasis, which correlate with disease progression and decreased survival times. The final section of the review addresses, firstly, the role of cancer-related platelet activation and thrombosis in the pathogenesis of secondary cardiovascular disorders and the associated mortality, particularly in lung cancer, which is second only to disease progression; secondly, the review addresses the potential role of antiplatelet agents in the adjunctive therapy of cancer.


Subject(s)
Cell-Derived Microparticles , Lung Neoplasms , Thrombosis , Humans , Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Thrombosis/metabolism , Lung Neoplasms/metabolism , Carcinogenesis/metabolism , Disease Progression
20.
Nanoscale Horiz ; 8(8): 1034-1042, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37435728

ABSTRACT

Extracellular vesicles (EVs), including nanoscale exosomes and ectosomes, hold promise as biomarkers that provide information about the cell of origin through their cargo of nucleic acids and proteins, both on their surface and within. Here, we develop a detection method of EVs based on light-induced acceleration of specific binding between their surface and antibody-modified microparticles, using a controlled microflow with three-dimensional analysis by confocal microscopy. Our method successfully detected 103-104 nanoscale EVs in liquid samples as small as a 500 nanoliters within 5 minutes, with the ability to distinguish multiple membrane proteins. Remarkably, we achieved the specific detection of EVs secreted from living cancer cell lines with high linearity, without the need for a time-consuming ultracentrifugation process that can take several hours. Furthermore, the detection range can be controlled by adjusting the action range of optical force using a defocused laser, consistent with the theoretical calculations. These findings demonstrate an ultrafast, sensitive, and quantitative approach for measuring biological nanoparticles, enabling innovative analyses of cell-to-cell communication and early diagnosis of various diseases, including cancer.


Subject(s)
Cell-Derived Microparticles , Exosomes , Extracellular Vesicles , Neoplasms , Humans , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Antibodies/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...