1
|
Ruiz-May E, Bojórquez-Velázquez E, Carrasco-Vargas H, Soto-Alvarez S, Rangel-Cova LS, Vargas-Hernández MA, Elizalde-Contreras JM, Torres De La Cruz VM, Vilchis-Ordoñez A, Palomo-Colli MÁ, Floriano TA, Ortega IP, Pelayo-Camacho R, Huerta-Nuñez LFE. Comparative proteomics uncovers vital molecular players of central nervous system leukemia among Mexican pediatric patients. J Proteomics 2025; 318:105458. [PMID: 40389027 DOI: 10.1016/j.jprot.2025.105458] [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: 11/08/2024] [Revised: 05/08/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric hematologic malignancy. The leukemic cells start in the bone marrow and spread to other vital organs and tissues, including the central nervous system (CNS). Hispanic males are the community with the highest incidence of ALL. Unfortunately, there is limited information on the molecular signatures of leukemic cell infiltration into the CNS, which marks the progression from ALL to central nervous system leukemia (CNSL). To address this knowledge gap, we performed a label-free shotgun proteomics between the cerebrospinal fluid (CSF) from patients with ALL compared to those with confirmed CNSL. We were able to identify 619 proteins, including 340 proteins that had not been reported prevously in the CSF of subjects with CNSL. The major features associated with CNSL included the over-accumulation of cell-adhesion proteins such as L-selectin and several cadherin proteins, along with the modulation of enzymes associated with protein glycosylation, which may provide the appropriate conditions for leukemic cell infiltration into the CNS. The over-accumulation of proteins associated with ROS scavenging processes reveals hypoxic and redox homeostasis microenvironment conditions that favors ALL progression to CNSL. The further characterization of key differential proteins will be essential for establishing reliable molecular markers for CSNL. BIOLOGICAL SIGNIFICANCE: Central nervous system leukemia (CNSL) is a catastrophic progression of ALL defined by leukemic cells reaching the CNS. Limited comparative proteomics studies have been conducted previously between patients with ALL versus CNSL. While these studies were important pioneering steps, they were hindered by difficulties in obtaining the proper amount of protein samples related to CSF. They were based on minimal study subjects, and their proteomics pipelines did not include the depletion of high-abundance proteins. Defining the manners in which the CNSL proteome is perturbed compared to the ALL condition is paramount to understanding the molecular foundations of CNSL and identifying and characterizing protein markers for proper diagnosis. Improving the sensitivity and reliability of diagnosis using molecular markers would help clinicians to maximize patient outcomes and quality of life by implementing aggressive CNSL treatments as soon as possible in cases where infiltration has occurred, while avoiding exposure to toxic chemotherapies in patients where they are not yet necessary.
Collapse
Affiliation(s)
- Eliel Ruiz-May
- Instituto de Ecología A.C. (INECOL), Carretera Antigua a Coatepec No. 351, Congregación el Haya, CP 91073, Xalapa, Veracruz, Mexico.
| | - Esaú Bojórquez-Velázquez
- Instituto de Ecología A.C. (INECOL), Carretera Antigua a Coatepec No. 351, Congregación el Haya, CP 91073, Xalapa, Veracruz, Mexico
| | - Humberto Carrasco-Vargas
- Secretaría de la Defensa Nacional, Universidad del Ejército y Fuerza Aérea, Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicana, Escuela Militar de Graduados de Sanidad, México City, Mexico
| | - Silverio Soto-Alvarez
- Secretaría de la Defensa Nacional, Universidad del Ejército y Fuerza Aérea, Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicana, Escuela Militar de Graduados de Sanidad, México City, Mexico
| | - Laura S Rangel-Cova
- Secretaría de la Defensa Nacional, Universidad del Ejército y Fuerza Aérea, Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicana, Escuela Militar de Graduados de Sanidad, México City, Mexico
| | - Marco Antonio Vargas-Hernández
- Secretaría de la Defensa Nacional, Universidad del Ejército y Fuerza Aérea, Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicana, Escuela Militar de Graduados de Sanidad, México City, Mexico
| | - José M Elizalde-Contreras
- Instituto de Ecología A.C. (INECOL), Carretera Antigua a Coatepec No. 351, Congregación el Haya, CP 91073, Xalapa, Veracruz, Mexico
| | - Víctor M Torres De La Cruz
- Instituto Mexicano del Seguro Social, Centro de Investigación Biomédicas del Noreste, Monterrey, Nuevo León, Mexico
| | - Armando Vilchis-Ordoñez
- Hospital infantil de México Federico Goméz, Dr. Márquez 162 CP 06720, 4 Unidad de Educación e Investigación, Mexico
| | - Miguel Ángel Palomo-Colli
- Hospital infantil de México Federico Goméz, Dr. Márquez 162 CP 06720, 4 Unidad de Educación e Investigación, Mexico
| | - Tania Angeles Floriano
- Hospital infantil de México Federico Goméz, Dr. Márquez 162 CP 06720, 4 Unidad de Educación e Investigación, Mexico
| | - Israel Parra Ortega
- Hospital infantil de México Federico Goméz, Dr. Márquez 162 CP 06720, 4 Unidad de Educación e Investigación, Mexico
| | - Rosana Pelayo-Camacho
- Unidad de Educación e Investigación, IMSS/ Oncoinmunología de citómica del cáncer infantil, Mexico
| | - Lidia F E Huerta-Nuñez
- Secretaría de la Defensa Nacional, Universidad del Ejército y Fuerza Aérea, Centro de Investigación y Desarrollo del Ejército y Fuerza Aérea Mexicana, Escuela Militar de Graduados de Sanidad, México City, Mexico.
| |
Collapse
|
2
|
Chatterjee M, Gupta S, Kumar U, Parashar D, Maitra A, Das K. Extracellular vesicles in acute myeloid leukemia: The role in disease pathogenesis, potential biomarker, and application in clinical settings. Crit Rev Oncol Hematol 2025; 211:104743. [PMID: 40280220 DOI: 10.1016/j.critrevonc.2025.104743] [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: 03/27/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025] Open
Abstract
Acute myeloid leukemia (AML), the most prevalent type of blood cancer, is initiated in the bone marrow and eventually migrates into the blood. It accounts for a 5-year overall survival rate of 29.8 %. AML results from the formation of immature white blood cells, also called AML blasts, from hematopoietic stem cells which eventually give rise to abnormal white blood cells, termed AML cells. The interaction of AML cells with their microenvironment appears to be significantly important in the pathogenesis of AML. A growing body of evidence identifies extracellular vesicles (EVs) to be a key component in intercellular communication via the transfer of biomolecules, such as DNA, RNAs, proteins, non-coding RNAs, lipids, metabolites etc. Although the role of EVs in various solid tumors is well-established, EVs' contribution to the pathogenesis of blood cancer, such as AML remains ill-defined. The present review highlights how EVs promote the progression of AML by influencing leukemogenesis, survival, angiogenesis, chemotherapeutic resistance, and immune evasion. A significant number of EVs are found in the biofluids of AML patients which are shown to carry signature cargo molecules, thereby rendering the EVs as predictive biomarkers for AML pathogenesis. EV-based clinical trials are mentioned in the later part of the review. Finally, EV-based therapeutics and their limitations are also briefly discussed in the context of AML.
Collapse
Affiliation(s)
- Madhura Chatterjee
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India.
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India.
| | - Umesh Kumar
- Department of Biosciences, IMS Ghaziabad (University Courses Campus), NH9, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh 201015, India.
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Arindam Maitra
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India.
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India.
| |
Collapse
|
3
|
Forte D, Pellegrino RM, Falvo P, Garcia-Gonzalez P, Alabed HBR, Maltoni F, Lombardi D, Bruno S, Barone M, Pasini F, Fabbri F, Vannini I, Donati B, Cristiano G, Sartor C, Ronzoni S, Ciarrocchi A, Buratta S, Urbanelli L, Emiliani C, Soverini S, Catani L, Bertolini F, Argüello RJ, Cavo M, Curti A. Parallel single-cell metabolic analysis and extracellular vesicle profiling reveal vulnerabilities with prognostic significance in acute myeloid leukemia. Nat Commun 2024; 15:10878. [PMID: 39738118 PMCID: PMC11685939 DOI: 10.1038/s41467-024-55231-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive disease with a high relapse rate. In this study, we map the metabolic profile of CD34+(CD38low/-) AML cells and the extracellular vesicle signatures in circulation from AML patients at diagnosis. CD34+ AML cells display high antioxidant glutathione levels and enhanced mitochondrial functionality, both associated with poor clinical outcomes. Although CD34+ AML cells are highly dependent on glucose oxidation and glycolysis for energy, those from intermediate- and adverse-risk patients reveal increased mitochondrial dependence. Extracellular vesicles from AML are mainly enriched in stem cell markers and express antioxidant GPX3, with their profiles showing potential prognostic value. Extracellular vesicles enhance mitochondrial functionality and dependence on CD34+ AML cells via the glutathione/GPX4 axis. Notably, extracellular vesicles from adverse-risk patients enhance leukemia cell engraftment in vivo. Here, we show a potential noninvasive approach based on liquid 'cell-extracellular vesicle' biopsy toward a redefined metabolic stratification in AML.
Collapse
Affiliation(s)
- Dorian Forte
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences Section, University of Perugia, Perugia, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Paulina Garcia-Gonzalez
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Husam B R Alabed
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences Section, University of Perugia, Perugia, Italy
| | - Filippo Maltoni
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Davide Lombardi
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Samantha Bruno
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Martina Barone
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Federico Pasini
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ivan Vannini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Benedetta Donati
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Gianluca Cristiano
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Chiara Sartor
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Simona Ronzoni
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milano, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences Section, University of Perugia, Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences Section, University of Perugia, Perugia, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, Biochemical and Biotechnological Sciences Section, University of Perugia, Perugia, Italy
- Centro di Eccellenza sui Materiali Innovativi Nanostrutturati (CEMIN), University of Perugia, Perugia, Italy
| | - Simona Soverini
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
| | - Lucia Catani
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy
| | - Rafael José Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Michele Cavo
- Department of Medical and Surgical Sciences, Institute of Hematology "L. and A. Seràgnoli", University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy.
| |
Collapse
|
4
|
Lajevardi MS, Ashrafpour M, Mubarak SMH, Rafieyan B, Kiani A, Noori E, Roayaei Ardakani M, Montazeri M, Kouhi Esfahani N, Asadimanesh N, Khalili S, Payandeh Z. Dual roles of extracellular vesicles in acute lymphoblastic leukemia: implications for disease progression and theranostic strategies. Med Oncol 2024; 42:11. [PMID: 39572459 PMCID: PMC11582151 DOI: 10.1007/s12032-024-02547-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/24/2024]
Abstract
Acute Lymphoblastic Leukemia (ALL) is a heterogeneous blood cancer characterized by the uncontrolled growth of immature lymphoid cells due to dysregulated signaling pathways. It is the most common pediatric cancer, with high cure rates in children, but significantly lower survival rates in adults. Current theranostic strategies, including chemotherapy, immunotherapy, and nanomedicine, aim to improve detection and treatment precision but are limited by side effects, drug resistance, high costs, and stability issues. Notably, extracellular vesicles (EVs) offer a promising alternative, addressing these limitations through their natural biocompatibility and targeted delivery capabilities. EVs play a dual role in ALL: they contribute to leukemia progression by promoting tumor growth, immune suppression, and drug resistance via the transfer of oncogenic molecules, while also serving as valuable non-invasive biomarkers due to their specific miRNA and protein content. Their ability to deliver therapeutic agents directly to leukemic cells, combined with their stability and low immunogenicity, makes EVs a compelling tool for improving ALL treatments. Indeed, by targeting the molecular pathways influenced by EVs or leveraging them for drug delivery, innovative therapeutic strategies can be developed to enhance treatment outcomes and reduce side effects. Thus, EVs represent a promising frontier for advancing theranostic strategies in ALL, offering new opportunities to improve diagnosis and treatment while overcoming the limitations of traditional therapies. This review will explore the dual roles of EVs in ALL, addressing their contributions to disease progression and their potential as therapeutic agents and biomarkers for early diagnosis and targeted therapies.
Collapse
Affiliation(s)
- Mahya Sadat Lajevardi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Shaden M H Mubarak
- Department of Clinical Laboratory Science, Faculty of Pharmacy, University of Kufa, Kufa, Iraq
| | - Behnoosh Rafieyan
- School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Arash Kiani
- Student Research Committee, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Effat Noori
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran
| | | | - Maryam Montazeri
- Razi Clinical Researches Development, Mazandaran University of Medical Science, Sari, Iran
| | - Niloofar Kouhi Esfahani
- Faculty of Medicine, People's Friendship University of Russia (Rudn University), Moscow, Russia
| | - Naghmeh Asadimanesh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Shahid Rajaee Teacher Training University, Tehran, 1678815811, Iran.
| | - Zahra Payandeh
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, 41346, Gothenburg, Sweden.
| |
Collapse
|
5
|
Chow JTS, Salmena L. 'One way, or another, I'm gonna find ya': miR-221-3p finds its targets via small extracellular vesicles. Haematologica 2024; 109:3094-3096. [PMID: 38634128 PMCID: PMC11443405 DOI: 10.3324/haematol.2024.285257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024] Open
Affiliation(s)
| | - Leonardo Salmena
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
7
|
Licari E, Cricrì G, Mauri M, Raimondo F, Dioni L, Favero C, Giussani A, Starace R, Nucera S, Biondi A, Piazza R, Bollati V, Dander E, D'Amico G. ActivinA modulates B-acute lymphoblastic leukaemia cell communication and survival by inducing extracellular vesicles production. Sci Rep 2024; 14:16083. [PMID: 38992199 PMCID: PMC11239915 DOI: 10.1038/s41598-024-66779-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/03/2024] [Indexed: 07/13/2024] Open
Abstract
Extracellular vesicles (EVs) are a new mechanism of cellular communication, by delivering their cargo into target cells to modulate molecular pathways. EV-mediated crosstalk contributes to tumor survival and resistance to cellular stress. However, the role of EVs in B-cell Acute Lymphoblastic Leukaemia (B-ALL) awaits to be thoroughly investigated. We recently published that ActivinA increases intracellular calcium levels and promotes actin polymerization in B-ALL cells. These biological processes guide cytoskeleton reorganization, which is a crucial event for EV secretion and internalization. Hence, we investigated the role of EVs in the context of B-ALL and the impact of ActivinA on this phenomenon. We demonstrated that leukemic cells release a higher number of EVs in response to ActivinA treatment, and they can actively uptake EVs released by other B-ALL cells. Under culture-induced stress conditions, EVs coculture promoted cell survival in B-ALL cells in a dose-dependent manner. Direct stimulation of B-ALL cells with ActivinA or with EVs isolated from ActivinA-stimulated cells was even more effective in preventing cell death. This effect can be possibly ascribed to the increase of vesiculation and modifications of EV-associated microRNAs induced by ActivinA. These data demonstrate that ActivinA boosts EV-mediated B-ALL crosstalk, improving leukemia survival in stress conditions.
Collapse
Affiliation(s)
- Eugenia Licari
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Giulia Cricrì
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
- Paediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano, Italy
| | - Mario Mauri
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Francesca Raimondo
- Clinical Proteomics and Metabolomic Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Laura Dioni
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Chiara Favero
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Alice Giussani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Rita Starace
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Silvia Nucera
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Andrea Biondi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- Hematology Division and Bone Marrow Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Valentina Bollati
- EPIGET Lab, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- CRC, Center for Environmental Health, University of Milan, Milan, Italy
- Occupational Health Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, Milan, Italy
| | - Erica Dander
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy
| | - Giovanna D'Amico
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Via Pergolesi, 20900, Monza, Italy.
| |
Collapse
|
8
|
Amirpour M, Kuhestani-Dehaghi B, Kheyrandish S, Hajipirloo LK, Khaffafpour Z, Keshavarz F, Allahbakhshian-Farsani M. The impact of exosomes derived from B-cell acute lymphoblastic leukemia as a growth factor on bone marrow mesenchymal stromal cells. Mol Biol Rep 2024; 51:749. [PMID: 38874800 DOI: 10.1007/s11033-024-09674-4] [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: 03/23/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Background The incidence of various types of cancers, including leukemia, is on the rise and many challenges in both drug resistance and complications related to chemotherapy appeared. Recently, the development and application of extracellular vesicles (EV) such as exosomes in the management of cancers, especially leukemia, holds great significance. In this article, we extracted exosomes from NALM6 cells and assessed their regulatory effects on proliferation and apoptosis in mesenchymal stem cells (MSCs). Method and result We first verified the exosomes using various techniques, including flow cytometry, transient electron microscopy, dynamic light scattering (DLS), and BCA protein assay. Then MTT analysis and flowcytometry (apoptosis and cell cycle assay) besides gene expressions were employed to determine the state of MSC proliferations. The results indicated that exosome-specific pan markers like CD9, CD63, and CD81 were present. Through DLS, we found out that the mean size of the exosomes was 89.68 nm. The protein content was determined to be 956.292 µg/ml. Analysis of MTT, flow cytometry (cell cycle and apoptosis assay), and RT-qPCR showed that in the dose of 50 µg/ml the proliferation of MSCs was increased significantly (p-value < 0.05). Conclusion All these data showed that exosomes use several signaling pathways to increase the MSCs' proliferation and drug resistance, ultimately leading to high mortalities and morbidities of acute lymphoblastic leukemia.
Collapse
Affiliation(s)
- Mozhgan Amirpour
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bentolhoda Kuhestani-Dehaghi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Setare Kheyrandish
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laya Khodayi Hajipirloo
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Khaffafpour
- Pediatric Congenital Hematologic Disorders Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Keshavarz
- Department of Immunology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Allahbakhshian-Farsani
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Amin S, Massoumi H, Tewari D, Roy A, Chaudhuri M, Jazayerli C, Krishan A, Singh M, Soleimani M, Karaca EE, Mirzaei A, Guaiquil VH, Rosenblatt MI, Djalilian AR, Jalilian E. Cell Type-Specific Extracellular Vesicles and Their Impact on Health and Disease. Int J Mol Sci 2024; 25:2730. [PMID: 38473976 PMCID: PMC10931654 DOI: 10.3390/ijms25052730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Extracellular vesicles (EVs), a diverse group of cell-derived exocytosed particles, are pivotal in mediating intercellular communication due to their ability to selectively transfer biomolecules to specific cell types. EVs, composed of proteins, nucleic acids, and lipids, are taken up by cells to affect a variety of signaling cascades. Research in the field has primarily focused on stem cell-derived EVs, with a particular focus on mesenchymal stem cells, for their potential therapeutic benefits. Recently, tissue-specific EVs or cell type-specific extracellular vesicles (CTS-EVs), have garnered attention for their unique biogenesis and molecular composition because they enable highly targeted cell-specific communication. Various studies have outlined the roles that CTS-EVs play in the signaling for physiological function and the maintenance of homeostasis, including immune modulation, tissue regeneration, and organ development. These properties are also exploited for disease propagation, such as in cancer, neurological disorders, infectious diseases, autoimmune conditions, and more. The insights gained from analyzing CTS-EVs in different biological roles not only enhance our understanding of intercellular signaling and disease pathogenesis but also open new avenues for innovative diagnostic biomarkers and therapeutic targets for a wide spectrum of medical conditions. This review comprehensively outlines the current understanding of CTS-EV origins, function within normal physiology, and implications in diseased states.
Collapse
Affiliation(s)
- Sohil Amin
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Hamed Massoumi
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
- Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Deepshikha Tewari
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Arnab Roy
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Madhurima Chaudhuri
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Cedra Jazayerli
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Abhi Krishan
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Mannat Singh
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Mohammad Soleimani
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Emine E. Karaca
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
- Department of Ophthalmology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Arash Mirzaei
- Department of Ophthalmology, University of Medical Sciences, Farabi Eye Hospital, Tehran 13366 16351, Iran;
| | - Victor H. Guaiquil
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Mark I. Rosenblatt
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Ali R. Djalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.A.); (H.M.); (D.T.); (A.R.); (M.C.); (C.J.); (A.K.); (M.S.); (M.S.); (E.E.K.); (V.H.G.); (M.I.R.); (E.J.)
- Richard and Loan Hill Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
10
|
Salehi A. A novel therapeutic strategy: the significance of exosomal miRNAs in acute myeloid leukemia. Med Oncol 2024; 41:62. [PMID: 38253748 DOI: 10.1007/s12032-023-02286-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024]
Abstract
Acute myeloid leukemia (AML) is a fast-growing blood cancer that interferes with the normal growth of blood cells in the bone marrow and blood. It is characterized by its unpredictable outlook and high death rate. The main treatment for AML is chemotherapy, but this often results in drug resistance and the possibility of the disease returning. For this reason, new biomarkers are necessary to diagnose, predict, and treat this disease. Research has demonstrated that cells responsible for AML release exosomes that interact with the disease's microenvironment. These exosomes have significant roles in promoting leukemia growth, suppressing normal hematopoiesis, facilitating angiogenesis, and contributing to drug resistance in AML. Further investigations have shown that these exosomes contain miRNAs, which are transferred to target cells and have functional roles. Biomarkers are utilized to assess various aspects of tumor cell behavior, including proliferation, apoptosis, angiogenesis, changes in the microenvironment, transfer of drug resistance, and stability in serum and blood plasma. In this research, we showed that exosomal miRNAs and exosomes have the potential to be used as indicators for detecting various phases of AML and can aid in its medical treatment. Furthermore, they can be specifically targeted for therapeutic purposes in addressing this condition.
Collapse
Affiliation(s)
- Ali Salehi
- Department of Cellular and Molecular Biology, Faculty of New Science and Technology, Tehran Medical Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Rahmati A, Mafi A, Soleymani F, Babaei Aghdam Z, Masihipour N, Ghezelbash B, Asemi R, Aschner M, Vakili O, Homayoonfal M, Asemi Z, Sharifi M, Azadi A, Mirzaei H, Aghadavod E. Circular RNAs: pivotal role in the leukemogenesis and novel indicators for the diagnosis and prognosis of acute myeloid leukemia. Front Oncol 2023; 13:1149187. [PMID: 37124518 PMCID: PMC10140500 DOI: 10.3389/fonc.2023.1149187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological malignancy and affected patients have poor overall survival (OS) rates. Circular RNAs (circRNAs) are a novel class of non-coding RNAs (ncRNAs) with a unique loop structure. In recent years, with the development of high-throughput RNA sequencing, many circRNAs have been identified exhibiting either up-regulation or down-regulation in AML patients compared with healthy controls. Recent studies have reported that circRNAs regulate leukemia cell proliferation, stemness, and apoptosis, both positively and negatively. Additionally, circRNAs could be promising biomarkers and therapeutic targets in AML. In this study, we present a comprehensive review of the regulatory roles and potentials of a number of dysregulated circRNAs in AML.
Collapse
Affiliation(s)
- Atefe Rahmati
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Basic Sciences, Faculty of Medicine, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Firooze Soleymani
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Babaei Aghdam
- Imaging Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Niloufar Masihipour
- Department of Medicine, Lorestan University of Medical Science, Lorestan, Iran
| | - Behrooz Ghezelbash
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Homayoonfal
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Azadi
- Department of Internal Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
- *Correspondence: Abbas Azadi, ; Esmat Aghadavod, ; Hamed Mirzaei, ;
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
- Department of Clinical Biochemistry, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- *Correspondence: Abbas Azadi, ; Esmat Aghadavod, ; Hamed Mirzaei, ;
| |
Collapse
|
12
|
Extracellular Vesicles in Haematological Disorders: A Friend or a Foe? Int J Mol Sci 2022; 23:ijms231710118. [PMID: 36077514 PMCID: PMC9455998 DOI: 10.3390/ijms231710118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Extracellular vesicles (EVs) have emerged as important mediators of homeostasis, immune modulation and intercellular communication. They are released by every cell of the human body and accordingly detected in a variety of body fluids. Interestingly, their expression can be upregulated under various conditions, such as stress, hypoxia, irradiation, inflammation, etc. Their cargo, which is variable and may include lipids, proteins, RNAs and DNA, reflects that of the parental cell, which offers a significant diagnostic potential to EVs. In line with this, an increasing number of studies have reported the important contribution of cancer-derived EVs in altering the tumour microenvironment and allowing for cancer progression and metastasis. As such, cancer-derived EVs may be used to monitor the development and progression of disease and to evaluate the potential response to treatment, which has generated much excitement in the field of oncology and particularly in haemato-oncology. Finally, EVs are able to transfer their cargo to target cells, modifying the properties of the recipient cell, which offers great therapeutic potential for EVs (either by specific drug delivery or by delivery of siRNAs and other inhibitory proteins). In this manuscript, we review the potential diagnostic use and therapeutic options of EVs in the context of haematological malignancies.
Collapse
|