1
|
Vafaeizadeh M, Abroun S, Soufi Zomorrod M. Effect of human bone marrow mesenchymal stem cell-derived microvesicles on the apoptosis of the multiple myeloma cell line U266. J Cancer Res Clin Oncol 2024; 150:299. [PMID: 38850382 PMCID: PMC11162395 DOI: 10.1007/s00432-024-05822-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Microvesicles are membraned particles produced by different types of cells recently investigated for anticancer purposes. The current study aimed to investigate the effects of human bone marrow mesenchymal stem cell-derived microvesicles (BMSC-MVs) on the multiple myeloma cell line U266. BMSC-MVs were isolated from BMSCs via ultracentrifugation and characterized using transmission electron microscopy (TEM) and dynamic light scattering (DLS). U266 cells were treated with 15, 30, 60, and 120 µg/mL BMSC-MVs for three and seven days and the effects of treatment in terms of viability, cytotoxicity, and DNA damage were investigated via the MTT assay, lactate dehydrogenase (LDH) assay, and 8‑hydroxy-2'-deoxyguanosine (8‑OHdG) measurement, respectively. Moreover, the apoptosis rate of the U266 cells treated with 60 µg/mL BMSC-MVs was also assessed seven days following treatment via flow cytometry. Ultimately, the expression level of BCL2, BAX, and CCND1 by the U266 cells was examined seven days following treatment with 60 µg/mL BMSC-MVs using qRT-PCR. RESULTS BMSC-MVs had an average size of ~ 410 nm. According to the MTT and LDH assays, BMSC-MV treatment reduced the U266 cell viability and mediated cytotoxic effects against them, respectively. Moreover, elevated 8‑OHdG levels following BMSC-MV treatment demonstrated a dose-dependent increase of DNA damage in the treated cells. BMSC-MV-treated U266 cells also exhibited an increased apoptosis rate after seven days of treatment. The expression level of BCL2 and CCND1 decreased in the treated cells whereas the BAX expression demonstrated an incremental pattern. CONCLUSIONS Our findings accentuate the therapeutic benefit of BMSC-MVs against the multiple myeloma cell line U266 and demonstrate how microvesicles could be of therapeutic advantage. Future in vivo studies could further corroborate these findings.
Collapse
Affiliation(s)
- Mona Vafaeizadeh
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Abroun
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mina Soufi Zomorrod
- Department of Hematology and Cell Therapy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
2
|
Arif S, Richer M, Larochelle S, Moulin VJ. Microvesicles derived from dermal myofibroblasts modify the integrity of the blood and lymphatic barriers using distinct endocytosis pathways. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e151. [PMID: 38939570 PMCID: PMC11080715 DOI: 10.1002/jex2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 06/29/2024]
Abstract
Microvesicles (MVs) are a subtype of extracellular vesicles that can transfer biological information from their producer cells to target cells. This communication can in turn affect both normal and pathological processes. Mounting evidence has revealed that dermal wound myofibroblasts (Wmyo) produce MVs, which can transfer biomolecules impacting receptor cells such as human dermal microvascular endothelial cells (HDMECs). While the effects of MVs on HDMECs are generally well described in the literature, little is known about the transport of MVs across the HDMEC barrier, and their potential effect on the barrier integrity remains unknown. Here, we investigated these roles of Wmyo-derived MVs on two sub-populations of HDMECs, blood endothelial cells (BECs) and lymphatic endothelial cells (LECs). Using an in vitro model to mimic the endothelial barrier, we showed that MVs crossed the LEC barrier but not the BEC barrier. In addition, we demonstrated that MVs were able to influence the cell-cell junctions of HDMECs. Specifically, we observed that after internalization via the predominantly caveolin-dependent pathway, MVs induced the opening of junctions in BECs. Conversely, in LECs, MVs mainly use the macropinocytosis pathway and induce closure of these junctions. Moreover, proteins in the MV membrane were responsible for this effect, but not specifically those belonging to the VEGF family. Finally, we found that once the LEC barrier permeability was reduced by MV stimuli, MVs ceased to cross the barrier. Conversely, when the BEC barrier was rendered permeable following stimulation with MVs, they were subsequently able to cross the barrier via the paracellular pathway. Taken together, these results suggest that the study of Wmyo-derived MVs offers valuable insights into their interaction with the HDMEC barrier in the context of wound healing. They highlight the potential significance of these MVs in the overall process.
Collapse
Affiliation(s)
- Syrine Arif
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Megan Richer
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Sébastien Larochelle
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Véronique J. Moulin
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuebecCanada
| |
Collapse
|
3
|
Motallebzadeh Khanmiri J, Mousavi SH, Alizadeh S, Khani-Eshratabadi M. Microvesicles Derived from Human Placental Mesenchymal Stem Cells Induce Autophagy and Apoptosis in Acute Myeloid Leukemia. JOURNAL OF ADVANCES IN MEDICAL AND BIOMEDICAL RESEARCH 2023; 31:574-584. [DOI: 10.30699/jambs.31.149.574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
|
4
|
Ghaffari K, Moradi-Hasanabad A, Sobhani-Nasab A, Javaheri J, Ghasemi A. Application of cell-derived exosomes in the hematological malignancies therapy. Front Pharmacol 2023; 14:1263834. [PMID: 37745073 PMCID: PMC10515215 DOI: 10.3389/fphar.2023.1263834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 08/31/2023] [Indexed: 09/26/2023] Open
Abstract
Exosomes are small membrane vesicles of endocytic origin that are produced by both tumor and normal cells and can be found in physiological fluids like plasma and cell culture supernatants. They include cytokines, growth factors, proteins, lipids, RNAs, and metabolites and are important intercellular communication controllers in several disorders. According to a vast amount of research, exosomes could support or inhibit tumor start and diffusion in a variety of solid and hematological malignancies by paracrine signaling. Exosomes are crucial therapeutic agents for a variety of illnesses, such as cancer and autoimmune diseases. This review discusses the most current and encouraging findings from in vitro and experimental in vivo research, as well as the scant number of ongoing clinical trials, with a focus on the impact of exosomes in the treatment of malignancies. Exosomes have great promise as carriers of medications, antagonists, genes, and other therapeutic materials that can be incorporated into their core in a variety of ways. Exosomes can also alter the metabolism of cancer cells, alter the activity of immunologic effectors, and alter non-coding RNAs, all of which can alter the tumor microenvironment and turn it from a pro-tumor to an anti-tumor milieu. This subject is covered in the current review, which also looks at how exosomes contribute to the onset and progression of hematological malignancies, as well as their importance in diagnosing and treating these conditions.
Collapse
Affiliation(s)
- Kazem Ghaffari
- Department of Basic and Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran
| | - Amin Moradi-Hasanabad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Sobhani-Nasab
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Javaheri
- Department of Health and Community Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Ali Ghasemi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
5
|
Khani-Eshratabadi M, Mousavi SH, Zarrabi M, Motallebzadeh Khanmiri J, Zeinali Bardar Z. Human Umbilical Cord Mesenchymal Stem Cell-Derived Microvesicles Could Induce Apoptosis and Autophagy in Acute Myeloid Leukemia. IRANIAN BIOMEDICAL JOURNAL 2023; 27:247-56. [PMID: 37873637 PMCID: PMC10707811 DOI: 10.61186/ibj.27.5.247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/31/2023] [Indexed: 12/17/2023]
Abstract
Background Microvesicles (MV) have been identified as candidate biomarkers for treating acute myeloid leukemia (AML). This study investigated the effects of human umbilical cord-derived mesenchymal stem cell (hUCMSC)-derived MVs on apoptosis and autophagy in the KG-1 leukemic cell line. Methods The hUCMSCs were cultured and characterized by flow cytometry. MVs were isolated by ultracentrifugation, and the concentration was determined using the Bradford method. The characteristics of MVs were confirmed using transmission electron microscopy, flow cytometry, and dynamic light scattering methods. KG-1 cells were treated with the desired concentrations of MVs for 24 h. The apoptosis induction and reactive oxygen species production were evaluated using flow cytometry. RT-PCR was performed to evaluate apoptosis- and autophagy-related genes expression. Results Following tretment of KG-1 cells with 25, 50, and 100 μg/ml concentrations of MVs, the apoptosis rates were 47.85%, 47.15%, and 51.35% (p < 0.0001), and the autophagy-induced ROS levels were 73.9% (p < 0.0002), 84.8% (p < 0.0001), and 85.4% (p < 0.0001), respectively. BAX and ATG7 gene expression increased significantly at all concentrations compared to the control, and this level was higher at 50 μg/ml than that of the other concentrations. In addition, LC3 and Beclin 1 expression increased significantly in a concentration-dependen manner. Conversely, BCL2 expression decreased compared to the control. Conclusion Our findings indicate that hUCMSC-MVs could induce cell death pathways of autophagy and apoptosis in the KG-1 cell lines and exert potent antiproliferative and proapoptotic effects on KG-1 cells in vitro. Therefore, hUCMSC-MVs may be a potential approach for cancer therapy as a novel cell-to-cell communication strategy.
Collapse
Affiliation(s)
- Mohammad Khani-Eshratabadi
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Zarrabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Jamal Motallebzadeh Khanmiri
- Department of Hematology and Blood Transfusion Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zeinali Bardar
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Dangot A, Zavaro M, Bar-Lev TH, Bannon L, Zilberman A, Pickholz E, Avivi I, Aharon A. Characterization of extracellular vesicles in COVID-19 infection during pregnancy. Front Cell Dev Biol 2023; 11:1135821. [PMID: 37560162 PMCID: PMC10407400 DOI: 10.3389/fcell.2023.1135821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
Introduction: SARS-CoV-2 infection may cause a severe inflammatory response, inflicting severe morbidity and mortality. This risk is modestly increased in pregnant patients. Despite the hypercoagulability and immunosuppression associated with pregnancy, most pregnant women experience a mild COVID-19 infection. Maternal extracellular vesicles (EVs) may interact with endothelial and immune components to facilitate a favorable disease course. This pilot study aimed to explore the characteristics of EVs released during COVID-19 infection occurring during the third trimester of pregnancy. Methods: In this prospective study, blood samples were obtained from 16 healthy non-pregnant (NP), 18 healthy-pregnant (HP), and 22 COVID-19 positive pregnant subjects (CoV-P). Disease course and pregnancy outcomes were assessed and EVs were characterized. Of note, limited volumes of sample acquired from the subjects made it necessary to use smaller and different subsets of samples for each analysis. Results: The majority (91%) of the COVID-19-pregnant subjects (18 mild and 2 moderate disease) experienced good pregnancy-related outcomes. EV concentrations were higher in healthy-pregnant subjects compared to non-pregnant subjects (p = 0.0041) and lower in COVID-19-pregnant subjects compared to healthy-pregnant subjects (p = 0.0150). CD63 exosome marker expression was higher in EVs of healthy-pregnant subjects and COVID-19-pregnant subjects compared to EVs of non-pregnant subjects (p = 0.0149, p = 0.0028, respectively). Similar levels of SARS-CoV-2 entry proteins (ACE-2 and TMPRSS2) were found in all three groups. Cytokine content increased in healthy-pregnant subject-EVs compared to non-pregnant EVs, while IL-2 and IL-6 levels were decreased in COVID-19-pregnant subject-EVs compared to healthy-pregnant subject-EVs (p = 0.043, p = 0.0390, respectively). CD8+, cytotoxic T-cell marker, was lower in non-pregnant EVs compared to healthy-pregnant subject-EVs and to COVID-19-pregnant subjects (p = 0.0108, p < 0.0001, respectively). COVID-19- pregnant subject-EVs demonstrated higher levels of platelet activation marker (CD62P) than non-pregnant (p = 0.0327) and healthy-pregnant subjects (p = 0.0365). Endothelial marker EV-CD144+ was lower in healthy-pregnant subjects versus non-pregnant subjects (p = 0.0093), but similar in COVID-19-pregnant and non-pregnant subjects. Other EVs' coagulation markers/activity, D-Dimer and fibrinogen levels were similar in healthy-pregnant subjects and COVID-19 positive pregnant subjects. Conclusion: COVID-19 positive pregnant subjects' EVs demonstrated an attenuated inflammatory response, with no additional activation of the coagulation system.
Collapse
Affiliation(s)
- Ayelet Dangot
- Hematology Research Laboratory, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
- Obstetrics and Gynecology Department, Lis Hospital for Women, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mor Zavaro
- Hematology Research Laboratory, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Hana Bar-Lev
- Hematology Research Laboratory, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Lian Bannon
- Department of Medicine F, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ayala Zilberman
- Obstetrics and Gynecology Department, Lis Hospital for Women, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eliana Pickholz
- Hematology Research Laboratory, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Avivi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Hematology Department, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Anat Aharon
- Hematology Research Laboratory, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
7
|
Chang J, Yan S, Geng Z, Wang Z. Inhibition of splicing factors SF3A3 and SRSF5 contributes to As 3+/Se 4+ combination-mediated proliferation suppression and apoptosis induction in acute promyelocytic leukemia cells. Arch Biochem Biophys 2023; 743:109677. [PMID: 37356608 DOI: 10.1016/j.abb.2023.109677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/28/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
The low-dose combination of Arsenite (As3+) and selenite (Se4+) has the advantages of lower biological toxicity and better curative effects for acute promyelocytic leukemia (APL) therapy. However, the underlying mechanisms remain unclear. Here, based on the fact that the combination of 2 μM A3+ plus 4 μM Se4+ possessed a stronger anti-leukemic effect on APL cell line NB4 as compared with each individual, we employed iTRAQ-based quantitative proteomics to identify a total of 58 proteins that were differentially expressed after treatment with As3+/Se4+ combination rather than As3+ or Se4+ alone, the majority of which were involved in spliceosome pathway. Among them, eight proteins stood out by virtue of their splicing function and significant changes. They were validated as being decreased in mRNA and protein levels under As3+/Se4+ combination treatment. Further functional studies showed that only knockdown of two splicing factors, SF3A3 and SRSF5, suppressed the growth of NB4 cells. The reduction of SF3A3 was found to cause G1/S cell cycle arrest, which resulted in proliferation inhibition. Moreover, SRSF5 downregulation induced cell apoptosis through the activation of caspase-3. Taken together, these findings indicate that SF3A3 and SRSF5 function as pro-leukemic factors and can be potential novel therapeutic targets for APL.
Collapse
Affiliation(s)
- Jiayin Chang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China
| | - Shihai Yan
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China
| | - Zhirong Geng
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210046, PR China.
| | - Zhilin Wang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210023, PR China.
| |
Collapse
|
8
|
Zhou L, Shan Z, Fan J. Extracellular Vesicles Derived from Human Bone Marrow Stem Cells Inhibit Acute Lymphoblastic Leukemia Cell Growth by Inhibiting MAPK Pathway via the miR-29b-3p/GDF15 Axis. Acta Haematol 2022; 146:505-517. [PMID: 36327876 DOI: 10.1159/000527456] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/18/2022] [Indexed: 12/26/2023]
Abstract
INTRODUCTION Acute lymphoblastic leukemia (ALL) is a common hematologic neoplastic disease. This study discussed the effect of extracellular vesicles (EVs) released from bone marrow mesenchymal stem cells (BMSCs) on ALL cells and the mechanism. METHODS BMSCs-EVs were isolated by differential centrifugation and identified. The effect of BMSCs-EVs on ALL cell proliferation and apoptosis was evaluated. The expression of miR-29b-3p in ALL cells and EVs was detected. The uptake of EVs by ALL cells was observed. The effect of miR-29b-3p on ALL cell proliferation and apoptosis was assessed after silencing miR-29b-3p. The targeting relation of miR-29b-3p and GDF15 was analyzed by bioinformatics website and dual-luciferase assay. The role of GDF15 in proliferation and apoptosis of ALL cells was further confirmed, and Western blot assay was performed to measure MAPK pathway-related protein levels. RESULTS BMSC-derived EVs inhibited proliferation and promoted apoptosis of ALL cells, as shown by the up-regulation of caspase-3 and Bax expressions and down-regulation of Bcl-2 expression. EVs carried miR-29b-3p into ALL cells, upregulated miR-29b-3p expression in ALL cells, and inhibited GDF15 expression. Silencing of miR-29b-3p or overexpression of GDF15 partially reversed the effect of EVs. EVs inhibited the MAPK pathway through the miR-29b-3p/GDF15 axis. CONCLUSION BMSCs-EVs carried miR-29b-3p into ALL cells, upregulated miR-29b-3p, and inhibited GDF15 to suppress the MAPK pathway and further inhibit proliferation and promote apoptosis of ALL cells.
Collapse
Affiliation(s)
- Li Zhou
- Department of Hematopathology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Zhe Shan
- Department of Hematopathology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Jiangsha Fan
- Department of Hematopathology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| |
Collapse
|
9
|
Extracellular Vesicle-Mediated Mitochondrial Reprogramming in Cancer. Cancers (Basel) 2022; 14:cancers14081865. [PMID: 35454774 PMCID: PMC9032679 DOI: 10.3390/cancers14081865] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/01/2022] [Accepted: 04/02/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Mitochondria are important organelles involved in several key cellular processes including energy production and cell death regulation. For this reason, it is unsurprising that mitochondrial function and structure are altered in several pathological states including cancer. Cancer cells present variate strategies to generate sufficient energy to sustain their high proliferation rates. These adaptative strategies can be mediated by extracellular signals such as extracellular vesicles. These vesicles can alter recipient cellular behavior by delivering their molecular cargo. This review explores the different EV-mediated mitochondrial reprogramming mechanisms supporting cancer survival and progression. Abstract Altered metabolism is a defining hallmark of cancer. Metabolic adaptations are often linked to a reprogramming of the mitochondria due to the importance of these organelles in energy production and biosynthesis. Cancer cells present heterogeneous metabolic phenotypes that can be modulated by signals originating from the tumor microenvironment. Extracellular vesicles (EVs) are recognized as key players in intercellular communications and mediate many of the hallmarks of cancer via the delivery of their diverse biological cargo molecules. Firstly, this review introduces the most characteristic changes that the EV-biogenesis machinery and mitochondria undergo in the context of cancer. Then, it focuses on the EV-driven processes which alter mitochondrial structure, composition, and function to provide a survival advantage to cancer cells in the context of the hallmarks of cancers, such as altered metabolic strategies, migration and invasiveness, immune surveillance escape, and evasion of apoptosis. Finally, it explores the as yet untapped potential of targeting mitochondria using EVs as delivery vectors as a promising cancer therapeutic strategy.
Collapse
|
10
|
Allegra A, Petrarca C, Di Gioacchino M, Casciaro M, Musolino C, Gangemi S. Exosome-Mediated Therapeutic Strategies for Management of Solid and Hematological Malignancies. Cells 2022; 11:cells11071128. [PMID: 35406692 PMCID: PMC8997895 DOI: 10.3390/cells11071128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Exosomes are small membrane vesicles of endocytic origin containing cytokines, RNAs, growth factors, proteins, lipids, and metabolites. They have been identified as fundamental intercellular communication controllers in several diseases and an enormous volume of data confirmed that exosomes could either sustain or inhibit tumor onset and diffusion in diverse solid and hematological malignancies by paracrine signaling. Thus, exosomes might constitute a promising cell-free tumor treatment alternative. This review focuses on the effects of exosomes in the treatment of tumors, by discussing the most recent and promising data from in vitro and experimental in vivo studies and the few existing clinical trials. Exosomes are extremely promising as transporters of drugs, antagomir, genes, and other therapeutic substances that can be integrated into their core via different procedures. Moreover, exosomes can augment or inhibit non-coding RNAs, change the metabolism of cancer cells, and modify the function of immunologic effectors thus modifying the tumor microenvironment transforming it from pro-tumor to antitumor milieu. Here, we report the development of currently realized exosome modifiers that offer indications for the forthcoming elaboration of other more effective methods capable of enhancing the activity of the exosomes.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: (A.A.); (M.D.G.)
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Department of Medicine and Aging Sciences, G. D’Annunzio University, 66100 Chieti, Italy
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Correspondence: (A.A.); (M.D.G.)
| | - Marco Casciaro
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
11
|
Zhang L, Khadka B, Wu J, Feng Y, Long B, Xiao R, Liu J. Bone Marrow Mesenchymal Stem Cells-Derived Exosomal miR-425-5p Inhibits Acute Myeloid Leukemia Cell Proliferation, Apoptosis, Invasion and Migration by Targeting WTAP. Onco Targets Ther 2021; 14:4901-4914. [PMID: 34594112 PMCID: PMC8478487 DOI: 10.2147/ott.s286326] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 05/22/2021] [Indexed: 11/23/2022] Open
Abstract
Introduction Acute myeloid leukemia (AML) is a predominant blood malignancy with high mortality and severe morbidity. AML is affected by microRNAs (miRNAs) loaded in exosomes derived from bone marrow mesenchymal stem cells (BM-MSCs). MiR-425-5p has been reported to participate in different cancer models. However, the function of BM-MSCs-derived exosomal miR-425-5p in AML is unclear. Methods The expression of miR-425-5p was measured by qRT-PCR in clinical AML samples. The immunophenotype of BM-MSCs was analyzed using antibodies against CD44, CD90, and CD105. The exosome was isolated from BM-MSCs. The effect of BM-MSCs-derived exosomal miR-425-5p on AML was analyzed by CCK-8 assay, Edu assay, transwell assay, flow cytometry in AML cells. qRT-PCR, luciferase reporter gene assay and Western blot analysis were also conducted in AML cells. Results The expression levels of miR-425-5p were decreased in CD34 + CD38-AML cells from primary AML patients compared to that from the bone marrow of healthy cases, and were reduced in exosomes from AML patients compared that from healthy cases. Similarly, miR-425-5p was also down-regulated in AML cell lines compared with BM-MSCs. MiR-425-5p was able to express in exosomes from BM-MSCs. CCK-8, Edu, transwell assay and flow cytometry analysis revealed that BM-MSCs-derived exosomal miR-425-5p significantly inhibited cell viability, Edu positive cells, invasion and migration, and induced apoptosis of AML cells. Meanwhile, the expression levels of cleaved PARP and cleaved caspase3 were increased by BM-MSCs-derived exosomal miR-425-5p in cells. MiR-425-5p inhibited the expression of Wilms tumor 1-associated protein (WTAP). Moreover, overexpression of WTAP could reverse the miR-425-5p-induced inhibition effect on AML cell proliferation, apoptosis, migration and invasion. Conclusion BM-MSCs-derived exosomal miR-425-5p inhibits proliferation, invasion and migration of AML cells and induced apoptosis of AML cells by targeting WTAP. Therapeutically, BM-MSCs-derived exosomal miR-425-5p may serve as a potential target for AML therapy.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Bijay Khadka
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Jieying Wu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Yashu Feng
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Bing Long
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Ruozhi Xiao
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| | - Jiajun Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou City, Guangdong Province, 510630, People's Republic of China
| |
Collapse
|
12
|
Liu X, Yin W, Widjaya AS, Yang Y, Liu Y, Jiang Y. A practical strategy to subcutaneous administered in-situ gelling co-delivery system of arsenic and retinoic acid for the treatment of acute promyelocytic leukemia. Asian J Pharm Sci 2021; 16:633-642. [PMID: 34849168 PMCID: PMC8609443 DOI: 10.1016/j.ajps.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/10/2021] [Accepted: 07/03/2021] [Indexed: 11/20/2022] Open
Abstract
Arsenic trioxide (ATO) combined with all trans retinoic acid (ATRA) is the first choice for the treatment of low and medium risk acute promyelocytic leukemia (APL). Clinical studies reported that the combination of ATO and ATRA could achieve a significant curative effect. However, the retinoic acid syndrome, serious drug resistance and the short half-life in vivo which lead to frequent and large dose administration limit the application of ATRA. In addition, the preparations of arsenic are conventional injections and tablets in clinic, which has poor patients' compliance caused by frequent long-term administration and serious side effects. In order to overcome the above limitations, a phospholipid phase separation gel (PPSG) loaded with ATO and ATRA was developed. ATO+ATRA-PPSG (AAP), as a biodegradable sustained-release delivery system, was the first achievement of co-delivery of hydrophilic ATO and lipophilic ATRA with high drug loading which is the main problem in the application of nano preparation. The prepared PPSG displayed high safety and biocompatibility. The drug in PPSG was released slowly and continuously in vivo and in vitro for up to 10 d, which could reduce the side effects caused by the fluctuation of blood drug concentration and solve the problem of the long treatment cycle and frequent administration. In vivo pharmacokinetics depicted that PPSG could improve the bioavailability, decrease the peak concentration, and prolong the t1/2 of ATO and ATRA. Particularly, AAP significantly inhibited the tumor volume, extended the survival period of tumor-bearing mice, and promoted the differentiation of APL cells into normal cells. Therefore, ATO+ATRA-PPSG not only could co-load hydrophilic ATO and lipophilic ATRA according to the clinical dosage, but also possessed the sustained-release and long-acting treatment effect which was expected to reduce administration time and ameliorate compliance of patients. Thus, it had great potential for clinical transformation and application.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| | - Weiwei Yin
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| | - Andy Samuel Widjaya
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| | - Yueying Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| | - Yunhu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| | - Yanyan Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201023, China
| |
Collapse
|
13
|
Yousefnia S. Mechanistic effects of arsenic trioxide on acute promyelocytic leukemia and other types of leukemias. Cell Biol Int 2021; 45:1148-1157. [PMID: 33527587 DOI: 10.1002/cbin.11563] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/25/2020] [Accepted: 01/31/2021] [Indexed: 12/13/2022]
Abstract
Acute promyelocytic leukemia (APL), a subtype of acute myeloid leukemia characterized with a translocation between promyelocytic leukemia gene (PML) on chromosome 15 and retinoic acid receptor alpha gene (RARα) on chromosome 17. Transcription of this fusion gene results in PML/RARα fusion protein blocking expression of critical genes involved in differentiation of myeloid cells through interaction with RAR element. PML/RARα fusion protein prevents normal function of PML and RARα as well as inhibiting apoptosis. Arsenic trioxide (ATO) is an important agent for the treatment of relapsed and newly diagnosed APL. ATO induces apoptosis, autophagy, and partial cellular differentiation as well as inhibiting cell growth and angiogenesis. Recognition of signaling pathways and molecular mechanisms induced by ATO can be effective for discovering novel treatment strategies to target leukemia cells. Also, it can be developed for the treatment of a variety of cancer cells. This review provides a perspective on anticancerous effects of ATO on APL and leukemia cells.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
14
|
Extracellular vesicles (EVs): What we know of the mesmerizing roles of these tiny vesicles in hematological malignancies? Life Sci 2021; 271:119177. [PMID: 33577843 DOI: 10.1016/j.lfs.2021.119177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Cancer is a complex disease in which a bidirectional collaboration between malignant cells and surrounding microenvironment creates an appropriate platform which ultimately facilitates the progression of the disease. The discovery of extracellular vesicles (EVs) was a turning point in the modern era of cancer biology, as their importance in human malignancies has set the stage to widen research interest in the field of cell-to-cell communication. The implication in short- and long-distance interaction via horizontally transfer of cellular components, ranging from non-coding RNAs to functional proteins, as well as stimulating target cells receptors by the means of ligands anchored on their membrane endows these "tiny vesicles with giant impacts" with incredible potential to re-educate normal tissues, and thus, to re-shape the surrounding niche. In this review, we highlight the pathogenic roles of EVs in human cancers, with an extensive focus on the recent advances in hematological malignancies.
Collapse
|
15
|
Bernardi S, Farina M. Exosomes and Extracellular Vesicles in Myeloid Neoplasia: The Multiple and Complex Roles Played by These " Magic Bullets". BIOLOGY 2021; 10:biology10020105. [PMID: 33540594 PMCID: PMC7912829 DOI: 10.3390/biology10020105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/25/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Extracellular vesicles (EVs) are released by the majority of cell types and can be isolated from both cell cultures and body fluids. They are involved in cell-to-cell communication and may shuttle different messages (RNA, DNA, and proteins). These messages are known to influence the microenvironment of cells and their behavior. In recent years, some evidence about the involvement of EVs and exosomes, an EV subgroup, in immunomodulation, the transfer of disease markers, and the treatment of myeloid malignancies have been reported. Little is known about these vesicles in this particular setting of hematologic neoplasia; here, we summarize and critically review the available results, aiming to encourage further investigations. Abstract Extracellular vesicles (exosomes, in particular) are essential in multicellular organisms because they mediate cell-to-cell communication via the transfer of secreted molecules. They are able to shuttle different cargo, from nucleic acids to proteins. The role of exosomes has been widely investigated in solid tumors, which gave us surprising results about their potential involvement in pathogenesis and created an opening for liquid biopsies. Less is known about exosomes in oncohematology, particularly concerning the malignancies deriving from myeloid lineage. In this review, we aim to present an overview of immunomodulation and the microenvironment alteration mediated by exosomes released by malicious myeloid cells. Afterwards, we review the studies reporting the use of exosomes as disease biomarkers and their influence in response to treatment, together with the recent experiences that have focused on the use of exosomes as therapeutic tools. The further development of new technologies and the increased knowledge of biological (exosomes) and clinical (myeloid neoplasia) aspects are expected to change the future approaches to these malignancies.
Collapse
Affiliation(s)
- Simona Bernardi
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
- Correspondence: or ; Tel.: +39-0303998464
| | - Mirko Farina
- Department of Clinical and Experimental Sciences, University of Brescia, Bone Marrow Transplant Unit, ASST Spedali Civili, 25123 Brescia, Italy;
- Centro di Ricerca Emato-Oncologica AIL (CREA), ASST Spedali Civili, 25123 Brescia, Italy
| |
Collapse
|