1
|
Silva-Sousa T, Usuda JN, Al-Arawe N, Frias F, Hinterseher I, Catar R, Luecht C, Riesner K, Hackel A, Schimke LF, Dias HD, Filgueiras IS, Nakaya HI, Camara NOS, Fischer S, Riemekasten G, Ringdén O, Penack O, Winkler T, Duda G, Fonseca DLM, Cabral-Marques O, Moll G. The global evolution and impact of systems biology and artificial intelligence in stem cell research and therapeutics development: a scoping review. Stem Cells 2024; 42:929-944. [PMID: 39230167 DOI: 10.1093/stmcls/sxae054] [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: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Advanced bioinformatics analysis, such as systems biology (SysBio) and artificial intelligence (AI) approaches, including machine learning (ML) and deep learning (DL), is increasingly present in stem cell (SC) research. An approximate timeline on these developments and their global impact is still lacking. We conducted a scoping review on the contribution of SysBio and AI analysis to SC research and therapy development based on literature published in PubMed between 2000 and 2024. We identified an 8 to 10-fold increase in research output related to all 3 search terms between 2000 and 2021, with a 10-fold increase in AI-related production since 2010. Use of SysBio and AI still predominates in preclinical basic research with increasing use in clinically oriented translational medicine since 2010. SysBio- and AI-related research was found all over the globe, with SysBio output led by the (US, n = 1487), (UK, n = 1094), Germany (n = 355), The Netherlands (n = 339), Russia (n = 215), and France (n = 149), while for AI-related research the US (n = 853) and UK (n = 258) take a strong lead, followed by Switzerland (n = 69), The Netherlands (n = 37), and Germany (n = 19). The US and UK are most active in SCs publications related to AI/ML and AI/DL. The prominent use of SysBio in ESC research was recently overtaken by prominent use of AI in iPSC and MSC research. This study reveals the global evolution and growing intersection among AI, SysBio, and SC research over the past 2 decades, with substantial growth in all 3 fields and exponential increases in AI-related research in the past decade.
Collapse
Affiliation(s)
- Thayna Silva-Sousa
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Júlia Nakanishi Usuda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Nada Al-Arawe
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Francisca Frias
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
| | - Irene Hinterseher
- Department of Vascular Surgery, Universitätsklinikum Ruppin-Brandenburg, Medizinische Hochschule Branderburg Theodor Fontane, 16816 Neuruppin, Germany
- Fakultät für Gesundheitswissenschaften Brandenburg, Gemeinsame Fakultät der Universität Potsdam, der Medizinischen Hochschule Brandenburg Theodor Fontane, und der Brandenburgischen Technischen Universität Cottbus-Senftenberg, 14476 Potsdam, Germany
- Vascular Surgery, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Katarina Riesner
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Alexander Hackel
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Lena F Schimke
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
| | - Haroldo Dutra Dias
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | | | - Helder I Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
| | - Niels Olsen Saraiva Camara
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
| | - Stefan Fischer
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, University Medical Center Schleswig Holstein Campus Lübeck, 23538 Lübeck, Germany
| | - Olle Ringdén
- Division of Pediatrics, Department of CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Olaf Penack
- Department of Hematology, Oncology, and Tumorimmunology, Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Tobias Winkler
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Georg Duda
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
| | - Dennyson Leandro M Fonseca
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
| | - Otávio Cabral-Marques
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo (SP), Brazil
- Department of Immunology, Institute of Biomedical Sciences, USP, SP, Brazil
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), USP, SP, Brazil
- Department of Medicine, Division of Molecular Medicine, Laboratory of Medical Investigation 29, USP School of Medicine (USPM), São Paulo (SP), Brazil
- D'OR Institute Research and Education, SP, Brazil
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Charité Universitätzsmedizin, corporate member of Freie Universität Berlin, Humboldt Universität zu Berlin, and Berlin Institute of Health (BIH), 10117 Berlin, Germany
- Julius Wolff Institute (JWI), Charité Universitätzsmedizin, 10117 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätzsmedizin, 10117 Berlin, Germany
| |
Collapse
|
2
|
Chansaenroj J, Kornsuthisopon C, Chansaenroj A, Samaranayake LP, Fan Y, Osathanon T. Potential of Dental Pulp Stem Cell Exosomes: Unveiling miRNA-Driven Regenerative Mechanisms. Int Dent J 2024:S0020-6539(24)01488-6. [PMID: 39368923 DOI: 10.1016/j.identj.2024.08.019] [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: 07/11/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 10/07/2024] Open
Abstract
Human dental pulp stem cells (hDPSCs) have emerged as a promising resource in regenerative medicine due to their unique ability to secrete exosomes containing a diverse array of bioactive molecules, particularly microRNAs (miRNAs). These exosomes appear to be essential for stimulating regenerative mechanisms, especially those associated with stem cell pluripotency and tissue repair. However, several challenges such as cargo specificity and delivery efficiency need to be addressed to maximise the therapeutic potential of hDPSC-derived exosomes and miRNA-based therapies. This narrative review explores hDPSCs' potential in regenerative medicine by examining their role in tissue engineering, secretome composition, exosome function, exosomal miRNA in diverse models, and miRNA profiling. Therefore, it is imperative to sustain ongoing research on miRNA to advance clinical applications in the field of regenerative medicine and dentistry. A comprehensive understanding of the specific miRNA composition within hDPSC-derived exosomes is essential to elucidate their mechanistic roles in diverse disease states and to inform the development of innovative therapeutic strategies. These findings hold significant potential for the development of innovative regenerative therapies and emphasises the importance of establishing a strong connection between translational research discoveries and clinical applications. hDPSC-derived exosomes and miRNA-based therapies play a crucial role in immune modulation, regenerative dentistry, and tissue repair.
Collapse
Affiliation(s)
- Jira Chansaenroj
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chatvadee Kornsuthisopon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Ajjima Chansaenroj
- Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lakshman P Samaranayake
- Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Yi Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
3
|
Eschenhagen T, Weinberger F. Challenges and perspectives of heart repair with pluripotent stem cell-derived cardiomyocytes. NATURE CARDIOVASCULAR RESEARCH 2024; 3:515-524. [PMID: 39195938 DOI: 10.1038/s44161-024-00472-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/04/2024] [Indexed: 08/29/2024]
Abstract
Here we aim at providing a concise but comprehensive overview of the perspectives and challenges of heart repair with pluripotent stem cell-derived cardiomyocytes. This Review comes at a time when consensus has been reached about the lack of relevant proliferative capacity of adult mammalian cardiomyocytes and the lack of new heart muscle formation with autologous cell sources. While alternatives to cell-based approaches will be shortly summarized, the focus lies on pluripotent stem cell-derived cardiomyocyte repair, which entered first clinical trials just 2 years ago. In the view of the authors, these early trials are important but have to be viewed as early proof-of-concept trials in humans that will hopefully provide first answers on feasibility, safety and the survival of allogeneic pluripotent stem cell-derived cardiomyocyte in the human heart. Better approaches have to be developed to make this approach clinically applicable.
Collapse
Affiliation(s)
- Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Florian Weinberger
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
- Cardiovascular Regeneration Program, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
4
|
Bistué-Rovira À, Rico LG, Bardina J, Juncà J, Granada I, Bradford JA, Ward MD, Salvia R, Solé F, Petriz J. Persistence of Chronic Lymphocytic Leukemia Stem-like Populations under Simultaneous In Vitro Treatment with Curcumin, Fludarabine, and Ibrutinib: Implications for Therapy Resistance. Int J Mol Sci 2024; 25:1994. [PMID: 38396682 PMCID: PMC10888954 DOI: 10.3390/ijms25041994] [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: 11/20/2023] [Revised: 01/11/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Leukemic stem cells (LSCs) possess similar characteristics to normal hematopoietic stem cells, including self-renewal capacity, quiescence, ability to initiate leukemia, and drug resistance. These cells play a significant role in leukemia relapse, persisting even after apparent remission. LSCs were first described in 1994 by Lapidot et al. Although they have been extensively studied in acute leukemia, more LSC research is still needed in chronic lymphocytic leukemia (CLL) to understand if reduced apoptosis in mature cells should still be considered as the major cause of this disease. Here, we provide new evidence suggesting the existence of stem-like cell populations in CLL, which may help to understand the disease as well as to develop effective treatments. In this study, we identified a potential leukemic stem cell subpopulation using the tetraploid CLL cell line I83. This subpopulation is characterized by diploid cells that were capable of generating the I83 tetraploid population. Furthermore, we adapted a novel flow cytometry analysis protocol to detect CLL subpopulations with stem cell properties in peripheral blood samples and primary cultures from CLL patients. These cells were identified by their co-expression of CD19 and CD5, characteristic markers of CLL cells. As previously described, increased alkaline phosphatase (ALP) activity is indicative of stemness and pluripotency. Moreover, we used this method to investigate the potential synergistic effect of curcumin in combination with fludarabine and ibrutinib to deplete this subpopulation. Our results confirmed the effectiveness of this ALP-based analysis protocol in detecting and monitoring leukemic stem-like cells in CLL. This analysis also identified limitations in eradicating these populations using in vitro testing. Furthermore, our findings demonstrated that curcumin significantly enhanced the effects of fludarabine and ibrutinib on the leukemic fraction, exhibiting synergistic effects (combination drug index, CDI 0.97 and 0.37, respectively). Our results lend support to the existence of potential stem-like populations in CLL cell lines, and to the idea that curcumin could serve as an effective adjuvant in therapies aimed at eliminating these populations and improving treatment efficacy.
Collapse
Affiliation(s)
- Àngel Bistué-Rovira
- Departament de Farmacologia, Terapèutica i Toxicologia, Universitat Autònoma de Barcelona (UAB), 08193 Cerdanyola del Vallès, Spain;
| | - Laura G. Rico
- Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08916 Badalona, Spain; (L.G.R.); (R.S.)
| | - Jorge Bardina
- Vall d’Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain
| | - Jordi Juncà
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Badalona, Spain; (J.J.); (I.G.); (F.S.)
| | - Isabel Granada
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Badalona, Spain; (J.J.); (I.G.); (F.S.)
| | - Jolene A. Bradford
- Thermo Fisher Scientific, Fort Collins, CO 80524, USA; (J.A.B.); (M.D.W.)
| | - Michael D. Ward
- Thermo Fisher Scientific, Fort Collins, CO 80524, USA; (J.A.B.); (M.D.W.)
| | - Roser Salvia
- Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08916 Badalona, Spain; (L.G.R.); (R.S.)
| | - Francesc Solé
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, 08916 Badalona, Spain; (J.J.); (I.G.); (F.S.)
| | - Jordi Petriz
- Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08916 Badalona, Spain; (L.G.R.); (R.S.)
| |
Collapse
|
5
|
Ran J, Dziedzic A, Naser IH, Itumalla R, Gupta JK, Rustagi S, Satapathy P, Khatib MN, Gaidhane S, Zahiruddin QS, Gaidhane AM, Sah R. Efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy: An umbrella review of systematic reviews. Int J Surg 2024; 110:01279778-990000000-01029. [PMID: 38320100 PMCID: PMC11487037 DOI: 10.1097/js9.0000000000001142] [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: 12/19/2023] [Accepted: 01/25/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Stem cell therapy (SCT) has emerged as a potential therapeutic avenue, with various cell types being explored for their efficacy in treating DCM. However, the safety and efficacy of these therapies have been the subject of numerous systematic reviews. This umbrella review aims to consolidate the existing evidence on stem cell interventions for DCM, providing a comprehensive overview of the current research landscape. METHODS This review was conducted following the JBI and PRISMA guidelines. Systematic reviews and meta-analyses of randomized controlled trials (RCTs) evaluating the safety and efficacy of SCT for DCM were included. Outcomes such as 6MWT, LVEDD, LVEF, MACE, NYHA, and QoL, among others, were considered. A literature search was executed across databases like PubMed, Embase, Web of Science, and Cochrane Database up to October 07, 2023. The quality of the included reviews was assessed using the JBI Checklist for Systematic Reviews and Research Syntheses. Data synthesis was carried out in both narrative and tabular formats, with the GRADE criteria guiding the determination of evidence certainty. RESULTS Nine systematic reviews met the inclusion criteria. LVEF found to be significantly improved with SCT. LVEDD and LVEDV assessments yielded mixed results, with some reviews observing significant changes. LVESV showed consistent reductions across multiple studies. BNP concentrations post-interventions were explored in several studies, with mixed findings. Health-related quality of life (HRQL) showed varied results, with some studies noting improvements and others finding no significant differences. NYHA classifications and 6-MWT results indicated potential benefits from stem cell treatments. SCT was observed to be generally safe. The certainty of evidence was low or very low for most of outcomes. CONCLUSION SCT showed has shown promise in treating DCM, with many studies highlighting its safety and potential benefits. Nonetheless, the existing data has its limitations due to biases in the RCTs studies. To truly establish the benefits of SCT for DCM, future high quality RCTS, are crucial.
Collapse
Affiliation(s)
- Jun Ran
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Arkadiusz Dziedzic
- Department of Conservative Dentistry with Endodontics, Medical University of Silesia, Katowice, Poland
| | - Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Ramaiah Itumalla
- School of Management, The Apollo University, Chittoor, Andhra Pradesh
| | | | | | - Prakasini Satapathy
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai
| | | | - Shilpa Gaidhane
- One Health Centre (COHERD), Jawaharlal Nehru Medical College
| | - Quazi Syed Zahiruddin
- South Asia Infant Feeding Research Network (SAIFRN), Division of Evidence Synthesis, Global Consortium of Public Health and Research
| | - Abhay M Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health. Datta Meghe Institute of Higher Education, Wardha
| | - Ranjit Sah
- Department of Clinical Microbiology, DY Patil Medical College, Hospital and Research Centre, DY Patil Vidyapeeth, Pune, Maharashtra, India
- Tribhuvan University Teaching Hospital, Kathmandu 46000, Nepal
| |
Collapse
|
6
|
Buchlak QD, Esmaili N, Moore J. Opportunities for developing neural stem cell treatments for acute ischemic stroke: A systematic review and gap analysis. J Clin Neurosci 2024; 120:64-75. [PMID: 38199150 DOI: 10.1016/j.jocn.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Ischemic stroke is a leading cause of disability and death. Current treatments are limited. Stem cell therapy has been highlighted as a potentially effective treatment to mitigate damage and restore function, but efficacy results are mixed. This study aimed to systematically review the literature on stem cell therapies for early acute ischemic stroke; and identify opportunities for future research to facilitate the development of an effective stem cell-based treatment. Original research published within the last 10 years that focused on the evaluation of a stem cell-based treatment for acute ischemic stroke in adult patients or subjects was included. Risk of bias was assessed using the SYRCLE and Cochrane risk of bias tools for animal and human studies, respectively. 3,396 articles were screened, 58 full-text articles were reviewed and 33 met inclusion criteria. Many studies appeared to be at risk of bias. Study designs and results were heterogeneous. Most studies were preclinical and involved stem cell administration within 24 hours. Seven studies tested the effects of multiple administration timepoints and one investigated repeat dosing. Six studies were conducted in humans and stem cell administration ranged from 24 hours to 90 days post stroke. Most studies employed the use of mesenchymal stem cells. The most appropriate cell delivery method appeared to be intra-arterial. Evidence suggests that stem cell therapy may be associated with beneficial effects. A literature gap analysis identified numerous opportunities for treatment development.
Collapse
Affiliation(s)
- Quinlan D Buchlak
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia; School of Medicine, University of Notre Dame Australia, Sydney, NSW, Australia.
| | - Nazanin Esmaili
- School of Medicine, University of Notre Dame Australia, Sydney, NSW, Australia; Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Justin Moore
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia; Department of Surgery, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
7
|
Racine L, Paldi A. Understanding Cell Differentiation Through Single-Cell Approaches: Conceptual Challenges of the Systemic Approach. Methods Mol Biol 2024; 2745:163-176. [PMID: 38060185 DOI: 10.1007/978-1-0716-3577-3_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
The cells of a multicellular organism are derived from a single zygote and genetically almost identical. Yet, they are phenotypically very different. This difference is the result of a process commonly called cell differentiation. How the phenotypic diversity emerges during ontogenesis or regeneration is a central and intensely studied but still unresolved issue in biology. Cell biology is facing conceptual challenges that are frequently confused with methodological difficulties. How to define a cell type? What stability or change means in the context of cell differentiation and how to deal with the ubiquitous molecular variations seen in the living cells? What are the driving forces of the change? We propose to reframe the problem of cell differentiation in a systemic way by incorporating different theoretical approaches. The new conceptual framework is able to capture the insights made at different levels of cellular organization and considered previously as contradictory. It also provides a formal strategy for further experimental studies.
Collapse
Affiliation(s)
- Laëtitia Racine
- Ecole Pratique des Hautes Etudes, PSL Research University, St-Antoine Research Center, INSERM U938, Paris, France
| | - Andras Paldi
- Ecole Pratique des Hautes Etudes, PSL Research University, St-Antoine Research Center, INSERM U938, Paris, France.
| |
Collapse
|
8
|
Pezhouman A, Nguyen NB, Kay M, Kanjilal B, Noshadi I, Ardehali R. Cardiac regeneration - Past advancements, current challenges, and future directions. J Mol Cell Cardiol 2023; 182:75-85. [PMID: 37482238 DOI: 10.1016/j.yjmcc.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Cardiovascular disease is the leading cause of mortality and morbidity worldwide. Despite improvements in the standard of care for patients with heart diseases, including innovation in pharmacotherapy and surgical interventions, none have yet been proven effective to prevent the progression to heart failure. Cardiac transplantation is the last resort for patients with severe heart failure, but donor shortages remain a roadblock. Cardiac regenerative strategies include cell-based therapeutics, gene therapy, direct reprogramming of non-cardiac cells, acellular biologics, and tissue engineering methods to restore damaged hearts. Significant advancements have been made over the past several decades within each of these fields. This review focuses on the advancements of: 1) cell-based cardiac regenerative therapies, 2) the use of noncoding RNA to induce endogenous cell proliferation, and 3) application of bioengineering methods to promote retention and integration of engrafted cells. Different cell sources have been investigated, including adult stem cells derived from bone marrow and adipose cells, cardiosphere-derived cells, skeletal myoblasts, and pluripotent stem cells. In addition to cell-based transplantation approaches, there have been accumulating interest over the past decade in inducing endogenous CM proliferation for heart regeneration, particularly with the use of noncoding RNAs such as miRNAs and lncRNAs. Bioengineering applications have focused on combining cell-transplantation approaches with fabrication of a porous, vascularized scaffold using biomaterials and advanced bio-fabrication techniques that may offer enhanced retention of transplanted cells, with the hope that these cells would better engraft with host tissue to improve cardiac function. This review summarizes the present status and future challenges of cardiac regenerative therapies.
Collapse
Affiliation(s)
- Arash Pezhouman
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States
| | - Ngoc B Nguyen
- Baylor College of Medicine, Department of Internal Medicine, Houston, Texas 77030, United States
| | - Maryam Kay
- Department of Medicine, Division of Cardiology, University of California, Los Angeles, CA 90095, United States
| | - Baishali Kanjilal
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Iman Noshadi
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, United States
| | - Reza Ardehali
- Baylor College of Medicine, Department of Medicine, Division of Cardiology, Houston, Texas 77030, United States; Texas Heart Institute, Houston, Texas 77030, United States.
| |
Collapse
|
9
|
Saber M, Shekari F, Mousavi SA, Moini A, Miri MS, Esfandiari F. JAK/STAT3 pathway promotes proliferation of ovarian aggregate-derived stem cells in vitro. Exp Cell Res 2023:113689. [PMID: 37355151 DOI: 10.1016/j.yexcr.2023.113689] [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: 05/02/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND The accurate identification and isolation of ovarian stem cells from mammalian ovaries remain a major challenge because of the lack of specific surface markers and suitable in vitro culture systems. Optimized culture conditions for in vitro expansion of ovarian stem cells would allow for identifying requirements of these stem cells for proliferation and differentiation that would pave the way to uncover role of ovarian stem cells in ovarian pathophysiology. Here, we used three-dimensional (3D) aggregate culture system for enrichment of ovarian stem cells and named them aggregate-derived stem cells (ASCs). We hypothesized that mimicking the ovarian microenvironment in vitro by using an aggregate model of the ovary would provide a suitable niche for the isolation of ovarian stem cells from adult mouse and human ovaries and wanted to find out the main cellular pathway governing the proliferation of these stem cells. RESULTS We showed that ovarian aggregates take an example from ovary microenvironment in terms of expression of ovarian markers, hormone secretion and supporting the viability of the cells. We found that aggregates-derived stem cells proliferate in vitro as long-term while remained expression of germline markers. These ovarian stem cells differentiated to oocyte like cells in vitro spontaneously. Transplantation of these stem cells in to chemotherapy mouse ovary could restore ovarian structure. RNA-sequencing analysis revealed that interleukin6 is upregulated pathway in ovarian aggregate-derived stem cells. Our data showed that JAK/Stat3 signaling pathway which is activated downstream of IL6 is critical for ovarian stem cells proliferation. CONCLUSIONS We developed a platform that is highly reproducible for in vitro propagation of ovarian stem cells. Our study provides a primary insight into cellular pathway governing the proliferation of ovarian stem cells.
Collapse
Affiliation(s)
- Maryam Saber
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed-Ahmad Mousavi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ashraf Moini
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Gynecology and Obstetrics, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran; Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Monireh-Sadat Miri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fereshteh Esfandiari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
10
|
Lo HW, Tapinos N. Editorial: Epigenetics and cellular plasticity in glioblastoma. Front Oncol 2023; 13:1179214. [PMID: 37020873 PMCID: PMC10068962 DOI: 10.3389/fonc.2023.1179214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/22/2023] Open
Affiliation(s)
- Hui-Wen Lo
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, United States
| | - Nikos Tapinos
- Department of Neuroscience, Brown University, Providence, RI, United States
- Laboratory of Cancer Epigenetics and Plasticity, Department of Neuroscience, Brown University, Providence, RI, United States
- *Correspondence: Nikos Tapinos,
| |
Collapse
|
11
|
Calthorpe RJ, Poulter C, Smyth AR, Sharkey D, Bhatt J, Jenkins G, Tatler AL. Complex roles of TGF-β signaling pathways in lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L285-L296. [PMID: 36625900 PMCID: PMC9988523 DOI: 10.1152/ajplung.00106.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/11/2023] Open
Abstract
As survival of extremely preterm infants continues to improve, there is also an associated increase in bronchopulmonary dysplasia (BPD), one of the most significant complications of preterm birth. BPD development is multifactorial resulting from exposure to multiple antenatal and postnatal stressors. BPD has both short-term health implications and long-term sequelae including increased respiratory, cardiovascular, and neurological morbidity. Transforming growth factor β (TGF-β) is an important signaling pathway in lung development, organ injury, and fibrosis and is implicated in the development of BPD. This review provides a detailed account on the role of TGF-β in antenatal and postnatal lung development, the effect of known risk factors for BPD on the TGF-β signaling pathway, and how medications currently in use or under development, for the prevention or treatment of BPD, affect TGF-β signaling.
Collapse
Affiliation(s)
- Rebecca J Calthorpe
- Lifespan & Population Health, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Caroline Poulter
- Department of Pediatrics, Queens Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Alan R Smyth
- Lifespan & Population Health, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- NIHR Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| | - Don Sharkey
- Centre for Perinatal Research, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Jayesh Bhatt
- Department of Pediatrics, Queens Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Amanda L Tatler
- NIHR Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
12
|
The Genes-Stemness-Secretome Interplay in Malignant Pleural Mesothelioma: Molecular Dynamics and Clinical Hints. Int J Mol Sci 2023; 24:ijms24043496. [PMID: 36834912 PMCID: PMC9963101 DOI: 10.3390/ijms24043496] [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: 01/23/2023] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
MPM has a uniquely poor somatic mutational landscape, mainly driven by environmental selective pressure. This feature has dramatically limited the development of effective treatment. However, genomic events are known to be associated with MPM progression, and specific genetic signatures emerge from the exceptional crosstalk between neoplastic cells and matrix components, among which one main area of focus is hypoxia. Here we discuss the novel therapeutic strategies focused on the exploitation of MPM genetic asset and its interconnection with the surrounding hypoxic microenvironment as well as transcript products and microvesicles representing both an insight into the pathogenesis and promising actionable targets.
Collapse
|
13
|
Grossman Z, Meyerhans A, Bocharov G. An integrative systems biology view of host-pathogen interactions: The regulation of immunity and homeostasis is concomitant, flexible, and smart. Front Immunol 2023; 13:1061290. [PMID: 36761169 PMCID: PMC9904014 DOI: 10.3389/fimmu.2022.1061290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/28/2022] [Indexed: 01/26/2023] Open
Abstract
The systemic bio-organization of humans and other mammals is essentially "preprogrammed", and the basic interacting units, the cells, can be crudely mapped into discrete sets of developmental lineages and maturation states. Over several decades, however, and focusing on the immune system, we and others invoked evidence - now overwhelming - suggesting dynamic acquisition of cellular properties and functions, through tuning, re-networking, chromatin remodeling, and adaptive differentiation. The genetically encoded "algorithms" that govern the integration of signals and the computation of new states are not fully understood but are believed to be "smart", designed to enable the cells and the system to discriminate meaningful perturbations from each other and from "noise". Cellular sensory and response properties are shaped in part by recurring temporal patterns, or features, of the signaling environment. We compared this phenomenon to associative brain learning. We proposed that interactive cell learning is subject to selective pressures geared to performance, allowing the response of immune cells to injury or infection to be progressively coordinated with that of other cell types across tissues and organs. This in turn is comparable to supervised brain learning. Guided by feedback from both the tissue itself and the neural system, resident or recruited antigen-specific and innate immune cells can eradicate a pathogen while simultaneously sustaining functional homeostasis. As informative memories of immune responses are imprinted both systemically and within the targeted tissues, it is desirable to enhance tissue preparedness by incorporating attenuated-pathogen vaccines and informed choice of tissue-centered immunomodulators in vaccination schemes. Fortunately, much of the "training" that a living system requires to survive and function in the face of disturbances from outside or within is already incorporated into its design, so it does not need to deep-learn how to face a new challenge each time from scratch. Instead, the system learns from experience how to efficiently select a built-in strategy, or a combination of those, and can then use tuning to refine its organization and responses. Efforts to identify and therapeutically augment such strategies can take advantage of existing integrative modeling approaches. One recently explored strategy is boosting the flux of uninfected cells into and throughout an infected tissue to rinse and replace the infected cells.
Collapse
Affiliation(s)
- Zvi Grossman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow, Russia
- Institute of Computer Science and Mathematical Modeling, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
14
|
Banovic M, Poglajen G, Vrtovec B, Ristic A. Contemporary Challenges of Regenerative Therapy in Patients with Ischemic and Non-Ischemic Heart Failure. J Cardiovasc Dev Dis 2022; 9:jcdd9120429. [PMID: 36547426 PMCID: PMC9783726 DOI: 10.3390/jcdd9120429] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 10/26/2022] [Indexed: 12/02/2022] Open
Abstract
It has now been almost 20 years since first clinical trials of stem cell therapy for heart repair were initiated. While initial preclinical data were promising and suggested that stem cells may be able to directly restore a diseased myocardium, this was never unequivocally confirmed in the clinical setting. Clinical trials of cell therapy did show the process to be feasible and safe. However, the clinical benefits of this treatment modality in patients with ischemic and non-ischemic heart failure have not been consistently confirmed. What is more, in the rapidly developing field of stem cell therapy in patients with heart failure, relevant questions regarding clinical trials' protocol streamlining, optimal patient selection, stem cell type and dose, and the mode of cell delivery remain largely unanswered. Recently, novel approaches to myocardial regeneration, including the use of pluripotent and allogeneic stem cells and cell-free therapeutic approaches, have been proposed. Thus, in this review, we aim to outline current knowledge and highlight contemporary challenges and dilemmas in clinical aspects of stem cell and regenerative therapy in patients with chronic ischemic and non-ischemic heart failure.
Collapse
Affiliation(s)
- Marko Banovic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
- Correspondence: (M.B.); (G.P.)
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (M.B.); (G.P.)
| | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Arsen Ristic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
| |
Collapse
|
15
|
Bueno C, Blanquer M, García-Bernal D, Martínez S, Moraleda JM. Binucleated human bone marrow-derived mesenchymal cells can be formed during neural-like differentiation with independence of any cell fusion events. Sci Rep 2022; 12:20615. [PMID: 36450873 PMCID: PMC9712539 DOI: 10.1038/s41598-022-24996-8] [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: 07/30/2022] [Accepted: 11/23/2022] [Indexed: 12/09/2022] Open
Abstract
Although it has been reported that bone marrow-derived cells (BMDCs) can transdifferentiate into neural cells, the findings are considered unlikely. It has been argued that the rapid neural transdifferentiation of BMDCs reported in culture studies is actually due to cytotoxic changes induced by the media. While transplantation studies indicated that BMDCs can form new neurons, it remains unclear whether the underlying mechanism is transdifferentiation or BMDCs-derived cell fusion with the existing neuronal cells. Cell fusion has been put forward to explain the presence of gene-marked binucleated neurons after gene-marked BMDCs transplantation. In the present study, we demostrated that human BMDCs can rapidly adopt a neural-like morphology through active neurite extension and binucleated human BMDCs can form with independence of any cell fusion events. We also showed that BMDCs neural-like differentiation involves the formation of intermediate cells which can then redifferentiate into neural-like cells, redifferentiate back to the mesenchymal fate or even repeatedly switch lineages without cell division. Furthermore, we have discovered that nuclei from intermediate cells rapidly move within the cell, adopting different morphologies and even forming binucleated cells. Therefore, our results provide a stronger basis for rejecting the idea that BMDCs neural transdifferentiation is merely an artefact.
Collapse
Affiliation(s)
- Carlos Bueno
- grid.10586.3a0000 0001 2287 8496Medicine Department and Hematopoietic Transplant and Cellular Therapy Unit, Institute of Biomedical Research (IMIB), Faculty of Medicine, University of Murcia, 30120 Murcia, Spain
| | - Miguel Blanquer
- grid.10586.3a0000 0001 2287 8496Medicine Department and Hematopoietic Transplant and Cellular Therapy Unit, Institute of Biomedical Research (IMIB), Faculty of Medicine, University of Murcia, 30120 Murcia, Spain
| | - David García-Bernal
- grid.10586.3a0000 0001 2287 8496Medicine Department and Hematopoietic Transplant and Cellular Therapy Unit, Institute of Biomedical Research (IMIB), Faculty of Medicine, University of Murcia, 30120 Murcia, Spain ,grid.10586.3a0000 0001 2287 8496Biochemistry, Molecular Biology and Immunology Department, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain
| | - Salvador Martínez
- grid.26811.3c0000 0001 0586 4893Instituto de Neurociencias de Alicante (UMH-CSIC), Universidad Miguel Hernandez, 03550 San Juan, Alicante, Spain
| | - José M. Moraleda
- grid.10586.3a0000 0001 2287 8496Medicine Department and Hematopoietic Transplant and Cellular Therapy Unit, Institute of Biomedical Research (IMIB), Faculty of Medicine, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
16
|
Evan T, Wang VMY, Behrens A. The roles of intratumour heterogeneity in the biology and treatment of pancreatic ductal adenocarcinoma. Oncogene 2022; 41:4686-4695. [PMID: 36088504 PMCID: PMC9568427 DOI: 10.1038/s41388-022-02448-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
Intratumour heterogeneity (ITH) has become an important focus of cancer research in recent years. ITH describes the cellular variation that enables tumour evolution, including tumour progression, metastasis and resistance to treatment. The selection and expansion of genetically distinct treatment-resistant cancer cell clones provides one explanation for treatment failure. However, tumour cell variation need not be genetically encoded. In pancreatic ductal adenocarcinoma (PDAC) in particular, the complex tumour microenvironment as well as crosstalk between tumour and stromal cells result in exceptionally variable tumour cell phenotypes that are also highly adaptable. In this review we discuss four different types of phenotypic heterogeneity within PDAC, from morphological to metabolic heterogeneity. We suggest that these different types of ITH are not independent, but, rather, can inform one another. Lastly, we highlight recent findings that suggest how therapeutic efforts may halt PDAC progression by constraining cellular heterogeneity.
Collapse
Affiliation(s)
- Theodore Evan
- Cancer Stem Cell Laboratory, The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK
| | | | - Axel Behrens
- Cancer Stem Cell Laboratory, The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, SW3 6JB, UK.
- Department of Surgery and Cancer, Imperial College London, London, SW7 2AZ, UK.
- CRUK Convergence Science Centre, Imperial College London, SW7 2AZ, London, UK.
| |
Collapse
|
17
|
Endometrial stem/progenitor cells: Properties, origins, and functions. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
18
|
Ashammakhi N, GhavamiNejad A, Tutar R, Fricker A, Roy I, Chatzistavrou X, Hoque Apu E, Nguyen KL, Ahsan T, Pountos I, Caterson EJ. Highlights on Advancing Frontiers in Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:633-664. [PMID: 34210148 PMCID: PMC9242713 DOI: 10.1089/ten.teb.2021.0012] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/15/2021] [Indexed: 01/05/2023]
Abstract
The field of tissue engineering continues to advance, sometimes in exponential leaps forward, but also sometimes at a rate that does not fulfill the promise that the field imagined a few decades ago. This review is in part a catalog of success in an effort to inform the process of innovation. Tissue engineering has recruited new technologies and developed new methods for engineering tissue constructs that can be used to mitigate or model disease states for study. Key to this antecedent statement is that the scientific effort must be anchored in the needs of a disease state and be working toward a functional product in regenerative medicine. It is this focus on the wildly important ideas coupled with partnered research efforts within both academia and industry that have shown most translational potential. The field continues to thrive and among the most important recent developments are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies that warrant special attention. Developments in the aforementioned areas as well as future directions are highlighted in this article. Although several early efforts have not come to fruition, there are good examples of commercial profitability that merit continued investment in tissue engineering. Impact statement Tissue engineering led to the development of new methods for regenerative medicine and disease models. Among the most important recent developments in tissue engineering are the use of three-dimensional bioprinting, organ-on-a-chip, and induced pluripotent stem cell technologies. These technologies and an understanding of them will have impact on the success of tissue engineering and its translation to regenerative medicine. Continued investment in tissue engineering will yield products and therapeutics, with both commercial importance and simultaneous disease mitigation.
Collapse
Affiliation(s)
- Nureddin Ashammakhi
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan, USA
| | - Amin GhavamiNejad
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Rumeysa Tutar
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Annabelle Fricker
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Xanthippi Chatzistavrou
- Department of Chemical Engineering and Material Science, College of Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Ehsanul Hoque Apu
- Department of Bioengineering, Henry Samueli School of Engineering, University of California, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Taby Ahsan
- RoosterBio, Inc., Frederick, Maryland, USA
| | - Ippokratis Pountos
- Academic Department of Trauma and Orthopaedics, University of Leeds, Leeds, United Kingdom
| | - Edward J. Caterson
- Division of Plastic Surgery, Department of Surgery, Nemours/Alfred I. du Pont Hospital for Children, Wilmington, Delaware, USA
| |
Collapse
|
19
|
Chakrabarti S, Hoque M, Jamil NZ, Singh VJ, Pollacksmith D, Meer N, Pezzano MT. Bone Marrow-Derived Cells Contribute to the Maintenance of Thymic Stroma including TECs. J Immunol Res 2022; 2022:6061746. [PMID: 35528618 PMCID: PMC9076333 DOI: 10.1155/2022/6061746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/24/2022] [Indexed: 12/29/2022] Open
Abstract
In paradox to critical functions for T-cell selection and self-tolerance, the thymus undergoes profound age-associated atrophy and loss of T-cell function, further enhanced by cancer therapies. Identifying thymic epithelial progenitor populations capable of forming functional thymic tissue will be critical in understanding thymic epithelial cell (TEC) ontogeny and designing strategies to reverse involution. We identified a new population of progenitor cells, present in both the thymus and bone marrow (BM) of mice, that coexpress the hematopoietic marker CD45 and the definitive thymic epithelial marker EpCAM and maintain the capacity to form functional thymic tissue. Confocal analysis and qRT-PCR of sorted cells from both BM and thymus confirmed coexpression of CD45 and EpCAM. Grafting of C57BL/6 fetal thymi under the kidney capsule of H2BGFP transgenic mice revealed that peripheral CD45+ EpCAM+ GFP-expressing cells migrate into the developing thymus and contribute to both TECs and FSP1-expressing thymic stroma. Sorted BM-derived CD45+ EpCAM+ cells contribute to reaggregate thymic organ cultures (RTOCs) and differentiate into keratin and FoxN1-expressing TECs, demonstrating that BM cells can contribute to the maintenance of TEC microenvironments previously thought to be derived solely from endoderm. BM-derived CD45+ EpCAM+ cells represent a new source of progenitor cells that contribute to thymic homeostasis. Future studies will characterize the contribution of BM-derived CD45+ EpCAM+ TEC progenitors to distinct functional TEC microenvironments in both the steady-state thymus and under conditions of demand. Cell therapies utilizing this population may help counteract thymic involution in cancer patients.
Collapse
Affiliation(s)
- Shami Chakrabarti
- Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY 10016, USA
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Mohammed Hoque
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Nawshin Zara Jamil
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Varan J. Singh
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Daniel Pollacksmith
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Neelab Meer
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
| | - Mark T. Pezzano
- Department of Biology, City College of New York CUNY, New York, NY 10031, USA
- Program in Biology, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
20
|
Figiel-Dabrowska A, Krześniak NE, Noszczyk BH, Domańska-Janik K, Sarnowska A. Efficiency assessment of irrigation as an alternative method for improving the regenerative potential of nonhealing wounds. Wound Repair Regen 2022; 30:303-316. [PMID: 35384136 PMCID: PMC9321893 DOI: 10.1111/wrr.13013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/29/2022]
Abstract
The application of mesenchymal stem/stromal cells (MSC) in regenerative medicine offers hope for the effective treatment of incurable or difficult‐to‐heal diseases. However, it requires the development of unified protocols for both safe and efficient cell acquisition and clinical usage. The therapeutic effect of fat grafts (containing stem cells) in non‐healing wounds has been discussed in previous studies, although the application requires local or general anaesthesia. The treatment of MSC derived from adipose tissue (ASC) could be a less invasive method, and efficient delivery could lead to more favourable outcomes, which should encourage clinicians to use such therapeutic approaches more frequently. Therefore, the aim of this study was to optimise the methods of ASC isolation, culture and administration while maintaining their high survival, proliferation and colonisation potential. The ASC were isolated by an enzymatic method and were characterised according to International Society for Cellular Therapy and International Federation for Adipose Therapeutics and Science guidelines. To assess the opportunity to obtain a sufficient number of cells for transplantation, long‐term cell cultures in two oxygen concentrations (5% vs. 21%) were conducted. For these cultures, the population doubling time, the cumulative time for cell population doublings and the rate of cell senescence were estimated. In a developed and pre‐defined protocol, ASC can be efficiently cultured at physiological oxygen concentrations (5%), which leads to faster proliferation and slower cell senescence. Subsequently, to select the optimal and minimally invasive methods of ASC transplantation, direct cell application with an irrigator or with skin dressings was analysed. Our results confirmed that both the presented methods of cell application allow for the safe delivery of isolated ASC into wounds without losing their vitality. Cells propagated in the described conditions and applied in non‐invasive cell application (with an irrigation system and dressings) to treat chronic wounds can be a potential alternative or supplement to more invasive clinical approaches.
Collapse
Affiliation(s)
| | - Natalia E Krześniak
- Department of Plastic and Reconstructive Surgery, Centre of Postgraduate Medical Education, Prof. W. Orlowski Memorial Hospital, Warsaw, Poland
| | - Bartłomiej H Noszczyk
- Department of Plastic and Reconstructive Surgery, Centre of Postgraduate Medical Education, Prof. W. Orlowski Memorial Hospital, Warsaw, Poland
| | | | - Anna Sarnowska
- Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
21
|
Rinkevich B, Ballarin L, Martinez P, Somorjai I, Ben‐Hamo O, Borisenko I, Berezikov E, Ereskovsky A, Gazave E, Khnykin D, Manni L, Petukhova O, Rosner A, Röttinger E, Spagnuolo A, Sugni M, Tiozzo S, Hobmayer B. A pan-metazoan concept for adult stem cells: the wobbling Penrose landscape. Biol Rev Camb Philos Soc 2022; 97:299-325. [PMID: 34617397 PMCID: PMC9292022 DOI: 10.1111/brv.12801] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/17/2022]
Abstract
Adult stem cells (ASCs) in vertebrates and model invertebrates (e.g. Drosophila melanogaster) are typically long-lived, lineage-restricted, clonogenic and quiescent cells with somatic descendants and tissue/organ-restricted activities. Such ASCs are mostly rare, morphologically undifferentiated, and undergo asymmetric cell division. Characterized by 'stemness' gene expression, they can regulate tissue/organ homeostasis, repair and regeneration. By contrast, analysis of other animal phyla shows that ASCs emerge at different life stages, present both differentiated and undifferentiated phenotypes, and may possess amoeboid movement. Usually pluri/totipotent, they may express germ-cell markers, but often lack germ-line sequestering, and typically do not reside in discrete niches. ASCs may constitute up to 40% of animal cells, and participate in a range of biological phenomena, from whole-body regeneration, dormancy, and agametic asexual reproduction, to indeterminate growth. They are considered legitimate units of selection. Conceptualizing this divergence, we present an alternative stemness metaphor to the Waddington landscape: the 'wobbling Penrose' landscape. Here, totipotent ASCs adopt ascending/descending courses of an 'Escherian stairwell', in a lifelong totipotency pathway. ASCs may also travel along lower stemness echelons to reach fully differentiated states. However, from any starting state, cells can change their stemness status, underscoring their dynamic cellular potencies. Thus, vertebrate ASCs may reflect just one metazoan ASC archetype.
Collapse
Affiliation(s)
- Baruch Rinkevich
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Loriano Ballarin
- Department of BiologyUniversity of PadovaVia Ugo Bassi 58/BPadova35121Italy
| | - Pedro Martinez
- Departament de Genètica, Microbiologia i EstadísticaUniversitat de BarcelonaAv. Diagonal 643Barcelona08028Spain
- Institut Català de Recerca i Estudis Avançats (ICREA)Passeig Lluís Companys 23Barcelona08010Spain
| | - Ildiko Somorjai
- School of BiologyUniversity of St AndrewsSt Andrews, FifeKY16 9ST, ScotlandUK
| | - Oshrat Ben‐Hamo
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Ilya Borisenko
- Department of Embryology, Faculty of BiologySaint‐Petersburg State UniversityUniversity Embankment, 7/9Saint‐Petersburg199034Russia
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center GroningenAntonius Deusinglaan 1Groningen9713 AVThe Netherlands
| | - Alexander Ereskovsky
- Department of Embryology, Faculty of BiologySaint‐Petersburg State UniversityUniversity Embankment, 7/9Saint‐Petersburg199034Russia
- Institut Méditerranéen de Biodiversité et d'Ecologie marine et continentale (IMBE), Aix Marseille University, CNRS, IRD, Avignon UniversityJardin du Pharo, 58 Boulevard Charles LivonMarseille13007France
- Koltzov Institute of Developmental Biology of Russian Academy of SciencesUlitsa Vavilova, 26Moscow119334Russia
| | - Eve Gazave
- Université de Paris, CNRS, Institut Jacques MonodParisF‐75006France
| | - Denis Khnykin
- Department of PathologyOslo University HospitalBygg 19, Gaustad Sykehus, Sognsvannsveien 21Oslo0188Norway
| | - Lucia Manni
- Department of BiologyUniversity of PadovaVia Ugo Bassi 58/BPadova35121Italy
| | - Olga Petukhova
- Collection of Vertebrate Cell CulturesInstitute of Cytology, Russian Academy of SciencesTikhoretsky Ave. 4St. Petersburg194064Russia
| | - Amalia Rosner
- Israel Oceanographic & Limnological ResearchNational Institute of OceanographyPOB 9753, Tel ShikmonaHaifa3109701Israel
| | - Eric Röttinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN)Nice06107France
- Université Côte d'Azur, Federative Research Institute – Marine Resources (IFR MARRES)28 Avenue de ValroseNice06103France
| | - Antonietta Spagnuolo
- Department of Biology and Evolution of Marine OrganismsStazione Zoologica Anton DohrnVilla ComunaleNaples80121Italy
| | - Michela Sugni
- Department of Environmental Science and Policy (ESP)Università degli Studi di MilanoVia Celoria 26Milan20133Italy
| | - Stefano Tiozzo
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche‐sur‐mer (LBDV)06234 Villefranche‐sur‐MerVillefranche sur MerCedexFrance
| | - Bert Hobmayer
- Institute of Zoology and Center for Molecular Biosciences, University of InnsbruckTechnikerstrInnsbruck256020Austria
| |
Collapse
|
22
|
Martinez P, Ballarin L, Ereskovsky AV, Gazave E, Hobmayer B, Manni L, Rottinger E, Sprecher SG, Tiozzo S, Varela-Coelho A, Rinkevich B. Articulating the "stem cell niche" paradigm through the lens of non-model aquatic invertebrates. BMC Biol 2022; 20:23. [PMID: 35057814 PMCID: PMC8781081 DOI: 10.1186/s12915-022-01230-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Stem cells (SCs) in vertebrates typically reside in "stem cell niches" (SCNs), morphologically restricted tissue microenvironments that are important for SC survival and proliferation. SCNs are broadly defined by properties including physical location, but in contrast to vertebrates and other "model" organisms, aquatic invertebrate SCs do not have clearly documented niche outlines or properties. Life strategies such as regeneration or asexual reproduction may have conditioned the niche architectural variability in aquatic or marine animal groups. By both establishing the invertebrates SCNs as independent types, yet allowing inclusiveness among them, the comparative analysis will allow the future functional characterization of SCNs.
Collapse
Affiliation(s)
- P Martinez
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Av. Diagonal 643, 08028, Barcelona, Spain.
- Institut Català de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - L Ballarin
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - A V Ereskovsky
- Aix Marseille University, Avignon Université, CNRS, IRD, IMBE, Marseille, France
- St. Petersburg State University, Biological Faculty, Universitetskaya emb. 7/9, St. Petersburg, 199034, Russia
- N. K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Vavilova Street 26, Moscow, 119334, Russia
| | - E Gazave
- Université de Paris, CNRS, Institut Jacques Monod, F-75006, Paris, France
| | - B Hobmayer
- Department of Zoology and Center of Molecular Biosciences, University of Innsbruck, Technikerstr. 25, 6020, Innsbruck, Austria
| | - L Manni
- Department of Biology, University of Padova, Via U. Bassi 58/B, 35100, Padova, Italy
| | - E Rottinger
- Université Côte d'Azur, CNRS, INSERM, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
- Université Côte d'Azur, Federative Research Institute - Marine Resources (IFR MARRES), Nice, France
| | - S G Sprecher
- Department of Biology, University of Fribourg, Chemin du Musee 10, 1700, Fribourg, Switzerland
| | - S Tiozzo
- Sorbonne Université, CNRS, Laboratoire de Biologie du Développement de Villefranche-sur-mer (LBDV), Paris, France
| | - A Varela-Coelho
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Av. da República, 2780-157, Oeiras, Portugal
| | - B Rinkevich
- Israel Oceanography and Limnological Research, National Institute of Oceanography, Tel Shikmona, P.O. Box 8030, 31080, Haifa, Israel.
| |
Collapse
|
23
|
Bhalerao A, Raut S, Noorani B, Mancuso S, Cucullo L. Molecular Mechanisms of Multi-Organ Failure in COVID-19 and Potential of Stem Cell Therapy. Cells 2021; 10:2878. [PMID: 34831101 PMCID: PMC8616204 DOI: 10.3390/cells10112878] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/18/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023] Open
Abstract
As the number of confirmed cases and deaths occurring from Coronavirus disease 2019 (COVID-19) surges worldwide, health experts are striving hard to fully comprehend the extent of damage caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although COVID-19 primarily manifests itself in the form of severe respiratory distress, it is also known to cause systemic damage to almost all major organs and organ systems within the body. In this review, we discuss the molecular mechanisms leading to multi-organ failure seen in COVID-19 patients. We also examine the potential of stem cell therapy in treating COVID-19 multi-organ failure cases.
Collapse
Affiliation(s)
- Aditya Bhalerao
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| | - Behnam Noorani
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Salvatore Mancuso
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA
| |
Collapse
|
24
|
Li JM, Kim S, Zhang Y, Bian F, Hu J, Lu R, Pflugfelder SC, Chen R, Li DQ. Single-Cell Transcriptomics Identifies a Unique Entity and Signature Markers of Transit-Amplifying Cells in Human Corneal Limbus. Invest Ophthalmol Vis Sci 2021; 62:36. [PMID: 34297801 PMCID: PMC8300054 DOI: 10.1167/iovs.62.9.36] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Differentiated from adult stem cells (ASCs), transit-amplifying cells (TACs) play an important role in tissue homeostasis, development, and regeneration. This study aimed to characterize the gene expression profile of a candidate TAC population in limbal basal epithelial cells using single-cell RNA sequencing (scRNA-seq). Methods Single cells isolated from the basal corneal limbus were subjected to scRNA-seq using the 10x Genomics platform. Cell types were clustered by graph-based visualization methods and unbiased computational analysis. BrdU proliferation assays, immunofluorescent staining, and real-time reverse transcription quantitative polymerase chain reaction were performed using multiple culture models of primary human limbal epithelial cells to characterize the TAC pool. Results Single-cell transcriptomics of 16,360 limbal basal cells revealed 12 cell clusters. A unique cluster (3.21% of total cells) was identified as a TAC entity, based on its less differentiated progenitor status and enriched exclusive proliferation marker genes, with 98.1% cells in S and G2/M phases. The cell cycle-dependent genes were revealed to be largely enriched by the TAC population. The top genes were characterized morphologically and functionally at protein and mRNA levels. The specific expression patterns of RRM2, TK1, CENPF, NUSAP1, UBE2C, and CDC20 were well correlated in a time- and cycle-dependent manner with proliferation stages in the cell growth and regeneration models. Conclusions For the first time, to the best of our knowledge, we have identified a unique TAC entity and uncovered a group of cell cycle-dependent genes that serve as TAC signature markers. The findings provide insight into ASCs and TACs and lay the foundation for understanding corneal homeostasis and diseases.
Collapse
Affiliation(s)
- Jin-Miao Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Sangbae Kim
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Yun Zhang
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Fang Bian
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Jiaoyue Hu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Rong Lu
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Stephen C Pflugfelder
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| | - Rui Chen
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - De-Quan Li
- Ocular Surface Center, Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
25
|
Diaz-Navarro R, Urrútia G, Cleland JG, Poloni D, Villagran F, Acosta-Dighero R, Bangdiwala SI, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Cochrane Database Syst Rev 2021; 7:CD013433. [PMID: 34286511 PMCID: PMC8406792 DOI: 10.1002/14651858.cd013433.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Stem cell therapy (SCT) has been proposed as an alternative treatment for dilated cardiomyopathy (DCM), nonetheless its effectiveness remains debatable. OBJECTIVES To assess the effectiveness and safety of SCT in adults with non-ischaemic DCM. SEARCH METHODS We searched CENTRAL in the Cochrane Library, MEDLINE, and Embase for relevant trials in November 2020. We also searched two clinical trials registers in May 2020. SELECTION CRITERIA Eligible studies were randomized controlled trials (RCT) comparing stem/progenitor cells with no cells in adults with non-ischaemic DCM. We included co-interventions such as the administration of stem cell mobilizing agents. Studies were classified and analysed into three categories according to the comparison intervention, which consisted of no intervention/placebo, cell mobilization with cytokines, or a different mode of SCT. The first two comparisons (no cells in the control group) served to assess the efficacy of SCT while the third (different mode of SCT) served to complement the review with information about safety and other information of potential utility for a better understanding of the effects of SCT. DATA COLLECTION AND ANALYSIS Two review authors independently screened all references for eligibility, assessed trial quality, and extracted data. We undertook a quantitative evaluation of data using random-effects meta-analyses. We evaluated heterogeneity using the I² statistic. We could not explore potential effect modifiers through subgroup analyses as they were deemed uninformative due to the scarce number of trials available. We assessed the certainty of the evidence using the GRADE approach. We created summary of findings tables using GRADEpro GDT. We focused our summary of findings on all-cause mortality, safety, health-related quality of life (HRQoL), performance status, and major adverse cardiovascular events. MAIN RESULTS We included 13 RCTs involving 762 participants (452 cell therapy and 310 controls). Only one study was at low risk of bias in all domains. There were many shortcomings in the publications that did not allow a precise assessment of the risk of bias in many domains. Due to the nature of the intervention, the main source of potential bias was lack of blinding of participants (performance bias). Frequently, the format of the continuous data available was not ideal for use in the meta-analysis and forced us to seek strategies for transforming data in a usable format. We are uncertain whether SCT reduces all-cause mortality in people with DCM compared to no intervention/placebo (mean follow-up 12 months) (risk ratio (RR) 0.84, 95% confidence interval (CI) 0.54 to 1.31; I² = 0%; studies = 7, participants = 361; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection in people with DCM (data could not be pooled; studies = 7; participants = 361; very low-certainty evidence). We are uncertain whether SCT improves HRQoL (standardized mean difference (SMD) 0.62, 95% CI 0.01 to 1.23; I² = 72%; studies = 5, participants = 272; very low-certainty evidence) and functional capacity (6-minute walk test) (mean difference (MD) 70.12 m, 95% CI -5.28 to 145.51; I² = 87%; studies = 5, participants = 230; very low-certainty evidence). SCT may result in a slight functional class (New York Heart Association) improvement (data could not be pooled; studies = 6, participants = 398; low-certainty evidence). None of the included studies reported major adverse cardiovascular events as defined in our protocol. SCT may not increase the risk of ventricular arrhythmia (data could not be pooled; studies = 8, participants = 504; low-certainty evidence). When comparing SCT to cell mobilization with granulocyte-colony stimulating factor (G-CSF), we are uncertain whether SCT reduces all-cause mortality (RR 0.46, 95% CI 0.16 to 1.31; I² = 39%; studies = 3, participants = 195; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection (studies = 1, participants = 60; very low-certainty evidence). SCT may not improve HRQoL (MD 4.61 points, 95% CI -5.62 to 14.83; studies = 1, participants = 22; low-certainty evidence). SCT may improve functional capacity (6-minute walk test) (MD 140.14 m, 95% CI 119.51 to 160.77; I² = 0%; studies = 2, participants = 155; low-certainty evidence). None of the included studies reported MACE as defined in our protocol or ventricular arrhythmia. The most commonly reported outcomes across studies were based on physiological measures of cardiac function where there were some beneficial effects suggesting potential benefits of SCT in people with non-ischaemic DCM. However, it is unclear if this intermediate effects translates into clinical benefits for these patients. With regard to specific aspects related to the modality of cell therapy and its delivery, uncertainties remain as subgroup analyses could not be performed as planned, making it necessary to wait for the publication of several studies that are currently in progress before any firm conclusion can be reached. AUTHORS' CONCLUSIONS We are uncertain whether SCT in people with DCM reduces the risk of all-cause mortality and procedural complications, improves HRQoL, and performance status (exercise capacity). SCT may improve functional class (NYHA), compared to usual care (no cells). Similarly, when compared to G-CSF, we are also uncertain whether SCT in people with DCM reduces the risk of all-cause mortality although some studies within this comparison observed a favourable effect that should be interpreted with caution. SCT may not improve HRQoL but may improve to some extent performance status (exercise capacity). Very low-quality evidence reflects uncertainty regarding procedural complications. These suggested beneficial effects of SCT, although uncertain due to the very low certainty of the evidence, are accompanied by favourable effects on some physiological measures of cardiac function. Presently, the most effective mode of administration of SCT and the population that could benefit the most is unclear. Therefore, it seems reasonable that use of SCT in people with DCM is limited to clinical research settings. Results of ongoing studies are likely to modify these conclusions.
Collapse
Affiliation(s)
- Rienzi Diaz-Navarro
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Gerard Urrútia
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - John Gf Cleland
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Daniel Poloni
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Francisco Villagran
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Roberto Acosta-Dighero
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
- School of Physiotherapy, Faculty of Health Sciences, Universidad San Sebastian, Santiago, Chile
| | - Shrikant I Bangdiwala
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Gabriel Rada
- Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eva Madrid
- Interdisciplinary Centre for Health Studies CIESAL, Universidad de Valparaíso, Viña del Mar, Chile
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
26
|
Barua A, Beygi A, Hatzikirou H. Close to Optimal Cell Sensing Ensures the Robustness of Tissue Differentiation Process: The Avian Photoreceptor Mosaic Case. ENTROPY 2021; 23:e23070867. [PMID: 34356408 PMCID: PMC8303396 DOI: 10.3390/e23070867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/05/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022]
Abstract
The way that progenitor cell fate decisions and the associated environmental sensing are regulated to ensure the robustness of the spatial and temporal order in which cells are generated towards a fully differentiating tissue still remains elusive. Here, we investigate how cells regulate their sensing intensity and radius to guarantee the required thermodynamic robustness of a differentiated tissue. In particular, we are interested in finding the conditions where dedifferentiation at cell level is possible (microscopic reversibility), but tissue maintains its spatial order and differentiation integrity (macroscopic irreversibility). In order to tackle this, we exploit the recently postulated Least microEnvironmental Uncertainty Principle (LEUP) to develop a theory of stochastic thermodynamics for cell differentiation. To assess the predictive and explanatory power of our theory, we challenge it against the avian photoreceptor mosaic data. By calibrating a single parameter, the LEUP can predict the cone color spatial distribution in the avian retina and, at the same time, suggest that such a spatial pattern is associated with quasi-optimal cell sensing. By means of the stochastic thermodynamics formalism, we find out that thermodynamic robustness of differentiated tissues depends on cell metabolism and cell sensing properties. In turn, we calculate the limits of the cell sensing radius that ensure the robustness of differentiated tissue spatial order. Finally, we further constrain our model predictions to the avian photoreceptor mosaic.
Collapse
Affiliation(s)
- Arnab Barua
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Nöthnitzer Straße 46, 01062 Dresden, Germany; (A.B.); (A.B.)
| | - Alireza Beygi
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Nöthnitzer Straße 46, 01062 Dresden, Germany; (A.B.); (A.B.)
| | - Haralampos Hatzikirou
- Centre for Information Services and High Performance Computing, Technische Universität Dresden, Nöthnitzer Straße 46, 01062 Dresden, Germany; (A.B.); (A.B.)
- Mathematics Department, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence:
| |
Collapse
|
27
|
Greulich P, MacArthur BD, Parigini C, Sánchez-García RJ. Universal principles of lineage architecture and stem cell identity in renewing tissues. Development 2021; 148:269055. [PMID: 34100065 DOI: 10.1242/dev.194399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 04/01/2021] [Indexed: 01/20/2023]
Abstract
Adult tissues in multicellular organisms typically contain a variety of stem, progenitor and differentiated cell types arranged in a lineage hierarchy that regulates healthy tissue turnover. Lineage hierarchies in disparate tissues often exhibit common features, yet the general principles regulating their architecture are not known. Here, we provide a formal framework for understanding the relationship between cell molecular 'states' and cell 'types', based on the topology of admissible cell state trajectories. We show that a self-renewing cell type - if defined as suggested by this framework - must reside at the top of any homeostatic renewing lineage hierarchy, and only there. This architecture arises as a natural consequence of homeostasis, and indeed is the only possible way that lineage architectures can be constructed to support homeostasis in renewing tissues. Furthermore, under suitable feedback regulation, for example from the stem cell niche, we show that the property of 'stemness' is entirely determined by the cell environment, in accordance with the notion that stem cell identities are contextual and not determined by hard-wired, cell-intrinsic characteristics. This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Philip Greulich
- Mathematical Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.,Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK
| | - Ben D MacArthur
- Mathematical Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.,Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.,Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK.,The Alan Turing Institute, London, NW1 2DB, UK
| | - Cristina Parigini
- Mathematical Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.,Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK
| | - Rubén J Sánchez-García
- Mathematical Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK.,Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, UK
| |
Collapse
|
28
|
Differentiation of human adult-derived stem cells towards a neural lineage involves a dedifferentiation event prior to differentiation to neural phenotypes. Sci Rep 2021; 11:12034. [PMID: 34103613 PMCID: PMC8187441 DOI: 10.1038/s41598-021-91566-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Although it has been reported that mesenchymal stem cells isolated from adult tissues can be induced to overcome their mesenchymal fate and transdifferentiate into neural cells, the findings and their interpretation have been challenged. The main argument against this process is that the cells rapidly adopt neuron-like morphologies through retraction of the cytoplasm rather than active neurite extension. In this study, we examined the sequence of biological events during neural differentiation of human periodontal ligament-derived stem cells (hPDLSCs), human bone marrow-derived stem cells (hBMSCs) and human dental pulp-derived stem cells (hDPSCs) by time-lapse microscopy. We have demonstrated that hPDLSCs, hBMSCs and hDPSCs can directly differentiate into neuron-like cells without passing through a mitotic stage and that they shrink dramatically and change their morphology to that of neuron-like cells through active neurite extension. Furthermore, we observed micronuclei movement and transient cell nuclei lobulation concurrent to in vitro neurogenesis from hBMSCs and hDPSCs. Our results demonstrate that the differentiation of hPDLSCs, hBMSCs and hDPSCs towards a neural lineage occurs through a dedifferentiation step followed by differentiation to neural phenotypes, and therefore we definitively confirm that the rapid acquisition of the neural phenotype is via a differentiation trait.
Collapse
|
29
|
Aquaporins implicated in the cell proliferation and the signaling pathways of cell stemness. Biochimie 2021; 188:52-60. [PMID: 33894294 DOI: 10.1016/j.biochi.2021.04.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/11/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022]
Abstract
Aquaporins (AQPs) are water channel proteins facilitating passive transport of water and other small molecules across biomembranes. Regulation of osmotic homeostasis via AQPs is accompanied by dynamic participation of various cellular signaling pathways. Recently emerging evidence reveals that functional roles of AQPs are further extended from the osmotic regulation via water permeation into the cell proliferation and differentiation. In particular, anomalous expression of AQPs has been demonstrated in various types of cancer cells and cancer stem-like cells and it has been proposed as markers for proliferation and progression of cancer cells. Thus, a more comprehensive view on AQPs could bring a great interest in the cell stemness accompanied by the expression of AQPs. AQPs are broadly expressed across tissues and cells in a cell type- and lineage-specific manner during development via spatiotemporal transcriptional regulation. Moreover, AQPs are expressed in various adult stem cells and cells associated with a stem cell niche as well as cancer stem-like cells. However, the expression and regulatory mechanisms of AQP expression in stem cells have not been well understood. This review highlighted the AQPs expression in stem cell niches/stem cells and the involvement of AQPs in the cell proliferation and signaling pathways associated with cell stemness.
Collapse
|
30
|
Branch EA, Matuska AM, Plummer HA, Harrison RM, Anz AW. Platelet-Rich Plasma Devices Can Be Used to Isolate Stem Cells From Synovial Fluid at the Point of Care. Arthroscopy 2021; 37:893-900. [PMID: 33010328 DOI: 10.1016/j.arthro.2020.09.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/12/2020] [Accepted: 09/19/2020] [Indexed: 02/02/2023]
Abstract
PURPOSE To assess whether point-of-care devices designed for collecting cellular components from blood or bone marrow could be used to isolate viable stem cells from synovial fluid. METHODS Male and female patients older than 18 years old with either an acute, anterior cruciate ligament (ACL) injury or knee osteoarthritis (OA) with a minimum estimated 20 mL of knee effusion volunteered. Ten patients with an ACL injury and 10 patients with OA were enrolled. Two milliliters of collected synovial effusion were analyzed and cultured for cellular content. The remaining fluid was combined with whole blood and processed using a buffy-coat based platelet-rich plasma (PRP) processing system. Specimens were analyzed for cell counts, colony-forming unit (CFU) assays, differentiation assays, and flow cytometry. RESULTS ACL effusion fluid contained 42.1 ± 20.7 CFU/mL and OA effusion fluid contained 65.4 ± 42.1 CFU/mL. After PRP processing, the counts in ACL-PRP were 101.6 ± 66.1 CFU/mL and 114.8 ± 73.4 CFU/mL in the OA-PRP. Cells showed tri-lineage differentiation potential when cultured under appropriate parameters. When analyzed with flow cytometry, >95% of cells produced with culturing expressed cell surface markers typically expressed by known stem cell populations, specifically CD45-, CD73+, CD29+, CD44+, CD105+, and CD90+. CONCLUSIONS Multipotent viable stem cells can be harvested from knee synovial fluid, associated with an ACL injury or OA, and concentrated with a buffy coat-based PRP-processing device. CLINICAL RELEVANCE PRP devices can be used to harvest stem cells from effusion fluids. Methods to use effusion fluid associated with an ACL injury and OA should be investigated further.
Collapse
Affiliation(s)
- Eric A Branch
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | | | - Hillary A Plummer
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A
| | | | - Adam W Anz
- Andrews Research & Education Foundation, Gulf Breeze, Florida, U.S.A..
| |
Collapse
|
31
|
Pacheco-Herrero M, Soto-Rojas LO, Reyes-Sabater H, Garcés-Ramirez L, de la Cruz López F, Villanueva-Fierro I, Luna-Muñoz J. Current Status and Challenges of Stem Cell Treatment for Alzheimer's Disease. J Alzheimers Dis 2021; 84:917-935. [PMID: 34633316 PMCID: PMC8673502 DOI: 10.3233/jad-200863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases called tauopathies, such as Alzheimer's disease (AD), frontotemporal dementia, progressive supranuclear palsy, and Parkinson's disease, among others, are characterized by the pathological processing and accumulation of tau protein. AD is the most prevalent neurodegenerative disease and is characterized by two lesions: neurofibrillary tangles (NFTs) and neuritic plaques. The presence of NFTs in the hippocampus and neocortex in early and advanced stages, respectively, correlates with the patient's cognitive deterioration. So far, no drugs can prevent, decrease, or limit neuronal death due to abnormal pathological tau accumulation. Among potential non-pharmacological treatments, physical exercise has been shown to stimulate the development of stem cells (SCs) and may be useful in early stages. However, this does not prevent neuronal death from the massive accumulation of NFTs. In recent years, SCs therapies have emerged as a promising tool to repopulate areas involved in cognition in neurodegenerative diseases. Unfortunately, protocols for SCs therapy are still being developed and the mechanism of action of such therapy remains unclear. In this review, we show the advances and limitations of SCs therapy. Finally, we provide a critical analysis of its clinical use for AD.
Collapse
Affiliation(s)
- Mar Pacheco-Herrero
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Luis O. Soto-Rojas
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, State of Mexico, Mexico
| | - Heidy Reyes-Sabater
- Neuroscience Research Laboratory, Faculty of Health Sciences, Pontificia Universidad Católica Madre y Maestra, Dominican Republic
| | - Linda Garcés-Ramirez
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Fidel de la Cruz López
- Escuela Nacional de Ciencias Biológicas, Depto de Fisiología, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - José Luna-Muñoz
- National Dementia BioBank, Ciencias Biológicas, Facultad de Estudios Superiores Cuautitlán, UNAM, State of Mexico, Mexico
- Banco Nacional de Cerebros-UNPHU, Universidad Nacional Pedro Henríquez Ureña, Dominican Republic
| |
Collapse
|
32
|
Gao X, Cao Z. Gingiva-derived Mesenchymal Stem Cells and Their Potential Applications in Oral and Maxillofacial Diseases. Curr Stem Cell Res Ther 2020; 15:43-53. [PMID: 31702517 DOI: 10.2174/1574888x14666191107100311] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/02/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Stem cells are undifferentiated cells with multilineage differentiation potential. They can be collected from bone marrow, fat, amniotic fluid, and teeth. Stem cell-based therapies have been widely used to treat multiple diseases, such as cardiac disease, and hematological disorders. The cells may also be beneficial for controlling the disease course and promoting tissue regeneration in oral and maxillofacial diseases. Oral-derived gingival mesenchymal stem cells are easy to access and the donor sites heal rapidly without a scar. Such characteristics demonstrate the beneficial role of GMSCs in oral and maxillofacial diseases. OBJECTIVE We summarize the features of GMSCs, including their self-renewal, multipotent differentiation, immunomodulation, and anti-inflammation properties. We also discuss their applications in oral and maxillofacial disease treatment and tissue regeneration. CONCLUSION GMSCs are easily harvestable adult stem cells with outstanding proliferation, differentiation, and immunomodulation characteristics. A growing body of evidence indicates that GMSCs have strong potential use in accelerating wound healing and promoting the regeneration of bone defects, periodontium, oral neoplasms, salivary glands, peri-implantitis, and nerves. Moreover, alginate, polylactic acid and polycaprolactone can be used as biodegradable scaffolds for GMSC encapsulation. Various growth factors can be applied to the corresponding scaffolds to obtain the desired GMSC differentiation and phenotypes. Three-dimensional spheroid culture systems could optimize GMSC properties and improve the performance of the cells in tissue engineering. The immunomodulatory property of GMSCs in controlling oral and maxillofacial inflammation needs further research.
Collapse
Affiliation(s)
- Xudong Gao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST KLOS) & Key Laboratory for Oral Biomedical Engineering of Ministry of Education (KLOBME), School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Que WC, Qiu HQ, Chen HY, Wang SZ, Cheng Y, Liao LM. Systematic evaluation of therapeutic effects of stem cell transplantation trials for heart diseases in China. Clin Transplant 2020; 34:e14051. [PMID: 32946605 DOI: 10.1111/ctr.14051] [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: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND To systematically assess the quality of reports of clinical trials of stem cell for heart diseases published in Chinese. METHODS The quality of reports was assessed according to the CONSORT statement and the Jadad score. The association between the CONSORT scores and the reported therapeutic effects was evaluated. RESULTS A total of 36 randomized clinical trials were identified, and 1552 patients were included. The mean CONSORT score was 7.06 (SD = 2.99). The proportion of reports with a Jadad score of 3 was 8.33%. The improvement of left ventricular function, myocardial perfusion area, left ventricular diastolic diameter, and cardiac output decreased with the increase in the CONSORT score. CONCLUSIONS The percentages of high-quality reports published in Chinese on stem cell therapy for heart diseases are low. Although stem cell transplantation seems promising for heart diseases, high-quality studies are needed to verify the conclusions..
Collapse
Affiliation(s)
- Wan-Cai Que
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hong-Qiang Qiu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Hui-Yun Chen
- Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shao-Zhen Wang
- Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yu Cheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lian-Ming Liao
- Center of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
34
|
Ma SKY, Chan ASF, Rubab A, Chan WCW, Chan D. Extracellular Matrix and Cellular Plasticity in Musculoskeletal Development. Front Cell Dev Biol 2020; 8:781. [PMID: 32984311 PMCID: PMC7477050 DOI: 10.3389/fcell.2020.00781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Cellular plasticity refers to the ability of cell fates to be reprogrammed given the proper signals, allowing for dedifferentiation or transdifferentiation into different cell fates. In vitro, this can be induced through direct activation of gene expression, however this process does not naturally occur in vivo. Instead, the microenvironment consisting of the extracellular matrix (ECM) and signaling factors, directs the signals presented to cells. Often the ECM is involved in regulating both biochemical and mechanical signals. In stem cell populations, this niche is necessary for maintenance and proper function of the stem cell pool. However, recent studies have demonstrated that differentiated or lineage restricted cells can exit their current state and transform into another state under different situations during development and regeneration. This may be achieved through (1) cells responding to a changing niche; (2) cells migrating and encountering a new niche; and (3) formation of a transitional niche followed by restoration of the homeostatic niche to sequentially guide cells along the regenerative process. This review focuses on examples in musculoskeletal biology, with the concept of ECM regulating cells and stem cells in development and regeneration, extending beyond the conventional concept of small population of progenitor cells, but under the right circumstances even “lineage-restricted” or differentiated cells can be reprogrammed to enter into a different fate.
Collapse
Affiliation(s)
- Sophia Ka Yan Ma
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | | | - Aqsa Rubab
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Wilson Cheuk Wing Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,Department of Orthopedics Surgery and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
35
|
Piccoli M, Ghiroldi A, Monasky MM, Cirillo F, Ciconte G, Pappone C, Anastasia L. Reversine: A Synthetic Purine with a Dual Activity as a Cell Dedifferentiating Agent and a Selective Anticancer Drug. Curr Med Chem 2020; 27:3448-3462. [PMID: 30605049 DOI: 10.2174/0929867326666190103120725] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/27/2022]
Abstract
The development of new therapeutic applications for adult and embryonic stem cells has dominated regenerative medicine and tissue engineering for several decades. However, since 2006, induced Pluripotent Stem Cells (iPSCs) have taken center stage in the field, as they promised to overcome several limitations of the other stem cell types. Nonetheless, other promising approaches for adult cell reprogramming have been attempted over the years, even before the generation of iPSCs. In particular, two years before the discovery of iPSCs, the possibility of synthesizing libraries of large organic compounds, as well as the development of high-throughput screenings to quickly test their biological activity, enabled the identification of a 2,6-disubstituted purine, named reversine, which was shown to be able to reprogram adult cells to a progenitor-like state. Since its discovery, the effect of reversine has been confirmed on different cell types, and several studies on its mechanism of action have revealed its central role in inhibitory activity on several kinases implicated in cell cycle regulation and cytokinesis. These key features, together with its chemical nature, suggested a possible use of the molecule as an anti-cancer drug. Remarkably, reversine exhibited potent cytotoxic activity against several tumor cell lines in vitro and a significant effect in decreasing tumor progression and metastatization in vivo. Thus, 15 years since its discovery, this review aims at critically summarizing the current knowledge to clarify the dual role of reversine as a dedifferentiating agent and anti-cancer drug.
Collapse
Affiliation(s)
- Marco Piccoli
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Andrea Ghiroldi
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Michelle M Monasky
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Federica Cirillo
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Giuseppe Ciconte
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Carlo Pappone
- Arrhythmology Department, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy
| | - Luigi Anastasia
- Stem Cells for Tissue Engineering Lab, IRCCS Policlinico San Donato, piazza Malan 2, San Donato Milanese, Milan, Italy.,Department of Biomedical Sciences for Health, University of Milan, via Luigi Mangiagalli 31, 20133 Milan, Italy
| |
Collapse
|
36
|
Wolfien M, Klatt D, Salybekov AA, Ii M, Komatsu-Horii M, Gaebel R, Philippou-Massier J, Schrinner E, Akimaru H, Akimaru E, David R, Garbade J, Gummert J, Haverich A, Hennig H, Iwasaki H, Kaminski A, Kawamoto A, Klopsch C, Kowallick JT, Krebs S, Nesteruk J, Reichenspurner H, Ritter C, Stamm C, Tani-Yokoyama A, Blum H, Wolkenhauer O, Schambach A, Asahara T, Steinhoff G. Hematopoietic stem-cell senescence and myocardial repair - Coronary artery disease genotype/phenotype analysis of post-MI myocardial regeneration response induced by CABG/CD133+ bone marrow hematopoietic stem cell treatment in RCT PERFECT Phase 3. EBioMedicine 2020; 57:102862. [PMID: 32629392 PMCID: PMC7339012 DOI: 10.1016/j.ebiom.2020.102862] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 01/08/2023] Open
Abstract
Background Bone marrow stem cell clonal dysfunction by somatic mutation is suspected to affect post-infarction myocardial regeneration after coronary bypass surgery (CABG). Methods Transcriptome and variant expression analysis was studied in the phase 3 PERFECT trial post myocardial infarction CABG and CD133+ bone marrow derived hematopoetic stem cells showing difference in left ventricular ejection fraction (∆LVEF) myocardial regeneration Responders (n=14; ∆LVEF +16% day 180/0) and Non-responders (n=9; ∆LVEF -1.1% day 180/0). Subsequently, the findings have been validated in an independent patient cohort (n=14) as well as in two preclinical mouse models investigating SH2B3/LNK antisense or knockout deficient conditions. Findings 1. Clinical: R differed from NR in a total of 161 genes in differential expression (n=23, q<0•05) and 872 genes in coexpression analysis (n=23, q<0•05). Machine Learning clustering analysis revealed distinct RvsNR preoperative gene-expression signatures in peripheral blood acorrelated to SH2B3 (p<0.05). Mutation analysis revealed increased specific variants in RvsNR. (R: 48 genes; NR: 224 genes). 2. Preclinical:SH2B3/LNK-silenced hematopoietic stem cell (HSC) clones displayed significant overgrowth of myeloid and immune cells in bone marrow, peripheral blood, and tissue at day 160 after competitive bone-marrow transplantation into mice. SH2B3/LNK−/− mice demonstrated enhanced cardiac repair through augmenting the kinetics of bone marrow-derived endothelial progenitor cells, increased capillary density in ischemic myocardium, and reduced left ventricular fibrosis with preserved cardiac function. 3. Validation: Evaluation analysis in 14 additional patients revealed 85% RvsNR (12/14 patients) prediction accuracy for the identified biomarker signature. Interpretation Myocardial repair is affected by HSC gene response and somatic mutation. Machine Learning can be utilized to identify and predict pathological HSC response. Funding German Ministry of Research and Education (BMBF): Reference and Translation Center for Cardiac Stem Cell Therapy - FKZ0312138A and FKZ031L0106C, German Ministry of Research and Education (BMBF): Collaborative research center - DFG:SFB738 and Center of Excellence - DFG:EC-REBIRTH), European Social Fonds: ESF/IV-WM-B34-0011/08, ESF/IV-WM-B34-0030/10, and Miltenyi Biotec GmbH, Bergisch-Gladbach, Germany. Japanese Ministry of Health : Health and Labour Sciences Research Grant (H14-trans-001, H17-trans-002) Trial registration ClinicalTrials.gov NCT00950274
Collapse
Affiliation(s)
- Markus Wolfien
- Department of Systems Biology and Bioinformatics, University Rostock, Institute of Computer Science, Ulmenstrasse 69, 18057 Rostock, Germany.
| | - Denise Klatt
- Hannover Medical School, Institute of Experimental Hematology, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| | - Amankeldi A Salybekov
- Department of Advanced Medicine Science, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1143, Japan
| | - Masaaki Ii
- Nanobridge, LLC. 1-3-5-202, Sawaragi-Nishi Ibaraki Osaka 567-0868, Japan.
| | - Miki Komatsu-Horii
- Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | - Ralf Gaebel
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Philippou-Massier
- Ludwig-Maximilians-Universität München, LAFUGA Genomics, Gene Center, Feodor-Lynen-Strasse 25, 81377 Muenchen, Germany.
| | - Eric Schrinner
- University Medical Center Goettingen, Institute for Diagnostic and Interventional Radiology, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.
| | - Hiroshi Akimaru
- Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | - Erika Akimaru
- Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | - Robert David
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Jens Garbade
- Department of Cardiac Surgery, Heart Center University Medicine Leipzig, Strümpellstrasse 39, 04289 Leipzig, Germany.
| | - Jan Gummert
- Heart and diabetes center North Rhine Westfalia, University hospital of the Ruhr university Bochum, Georgstraße 11, 32545 Bad Oeynhausen, Germany.
| | - Axel Haverich
- Medical school Hannover, Department of heart-, thoracic- and vascular surgery, Carl Neuberg Strasse 1, 30625 Hannover, Germany.
| | - Holger Hennig
- Department of Systems Biology and Bioinformatics, University Rostock, Institute of Computer Science, Ulmenstrasse 69, 18057 Rostock, Germany.
| | - Hiroto Iwasaki
- Department of cardiothoracic surgery, Osaka city university, 1-4-3, Asahimachi, Abeno. Osaka, 545-8585. Japan.
| | - Alexander Kaminski
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Atsuhiko Kawamoto
- Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | - Christian Klopsch
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Johannes T Kowallick
- University Medical Center Goettingen, Institute for Diagnostic and Interventional Radiology, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.
| | - Stefan Krebs
- Ludwig-Maximilians-Universität München, LAFUGA Genomics, Gene Center, Feodor-Lynen-Strasse 25, 81377 Muenchen, Germany.
| | - Julia Nesteruk
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Hermann Reichenspurner
- Department of Cardiac and Vascular Surgery, University heart center Hamburg, Martinistraße. 52, 20246 Hamburg, Germany.
| | - Christian Ritter
- University Medical Center Goettingen, Institute for Diagnostic and Interventional Radiology, Robert-Koch-Strasse 40, 37075 Göttingen, Germany.
| | - Christof Stamm
- German Heart Center Berlin, Department of Heart-, Thoracic- and Vascular Surgery, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Ayumi Tani-Yokoyama
- Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| | - Helmut Blum
- Ludwig-Maximilians-Universität München, LAFUGA Genomics, Gene Center, Feodor-Lynen-Strasse 25, 81377 Muenchen, Germany.
| | - Olaf Wolkenhauer
- Department of Systems Biology and Bioinformatics, University Rostock, Institute of Computer Science, Ulmenstrasse 69, 18057 Rostock, Germany.
| | - Axel Schambach
- Hannover Medical School, Institute of Experimental Hematology, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| | - Takayuki Asahara
- Department of Advanced Medicine Science, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1143, Japan.
| | - Gustav Steinhoff
- Reference and Translation Center for Cardiac Stem Cell Therapy, Department Life, Light and Matter and Department of cardiac surgery, University Medicine Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
37
|
|
38
|
Grossman Z, Singh NJ, Simonetti FR, Lederman MM, Douek DC, Deeks SG. 'Rinse and Replace': Boosting T Cell Turnover To Reduce HIV-1 Reservoirs. Trends Immunol 2020; 41:466-480. [PMID: 32414695 DOI: 10.1016/j.it.2020.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/22/2022]
Abstract
Latent HIV-1 persists indefinitely during antiretroviral therapy (ART) as an integrated silent genome in long-lived memory CD4+ T cells. In untreated infections, immune activation increases the turnover of intrinsically long-lived provirus-containing CD4+ T cells. Those are 'washed out' as a result of their activation, which when coupled to viral protein expression can facilitate local inflammation and recruitment of uninfected cells to activation sites, causing latently infected cells to compete for survival. De novo infection can counter this washout. During ART, inflammation and CD4+ T cell activation wane, resulting in reduced cell turnover and a persistent reservoir. We propose accelerating reservoir washout during ART by triggering sequential waves of polyclonal CD4+ T cell activation while simultaneously enhancing virus protein expression. Reservoir reduction as an adjunct to other therapies might achieve lifelong viral control.
Collapse
Affiliation(s)
- Zvi Grossman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA; Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Nevil J Singh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Francesco R Simonetti
- 'L. Sacco' Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | | | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
39
|
Rico LG, Juncà J, Ward MD, Bradford JA, Petriz J. Flow cytometric significance of cellular alkaline phosphatase activity in acute myeloid leukemia. Oncotarget 2019; 10:6969-6980. [PMID: 31857851 PMCID: PMC6916750 DOI: 10.18632/oncotarget.27356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
In this prospective hospital-based cohort study that included 43 newly diagnosed patients with acute myeloid leukemia, flow cytometric cellular alkaline phosphatase (ALP) activity within primitive leukemic cells allowed us to identify two groups of patients at diagnosis according to the numbers of leukemic blasts expressing ≥ 12% of ALP+ cells (27 patients, Group A) and less than 12% of ALP+ cells (16 patients, Group B). Differences in outcome for complete response, relapse or treatment resistance, and exitus were statistically analyzed and were significant, when comparing the two groups. The overall survival (OS) and event-free survival (EFS) differences between Group A and B were statistically significant. The survival of Group A patients was significantly shorter than those for Group B. No significant relationship was detected in outcome when comparing ELN prognostic-risk group based on cytogenetic and molecular profile (patients in the favorable, intermediate, and adverse risk groups). Flow cytometric cellular ALP activity at diagnosis may be used to estimate relapses and disease persistence more accurately. The limitations of our study include the small number of patients enrolled and a short follow-up, due to its prospective nature.
Collapse
Affiliation(s)
- Laura G. Rico
- Functional Cytomics Group, Institut de Recerca contra la Leucèmia Josep Carreras, IJC Campus ICO-Germans Trias i Pujol, Institut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, UAB, Badalona, Spain
| | - Jordi Juncà
- Functional Cytomics Group, Institut de Recerca contra la Leucèmia Josep Carreras, IJC Campus ICO-Germans Trias i Pujol, Institut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, UAB, Badalona, Spain
- Institut Català d’Oncologia, Hospital Germans Trias i Pujol (HGTiP), Badalona, Spain
| | | | | | - Jordi Petriz
- Functional Cytomics Group, Institut de Recerca contra la Leucèmia Josep Carreras, IJC Campus ICO-Germans Trias i Pujol, Institut Germans Trias i Pujol (IGTP), Universitat Autònoma de Barcelona, UAB, Badalona, Spain
| |
Collapse
|
40
|
Grossman Z. Immunological Paradigms, Mechanisms, and Models: Conceptual Understanding Is a Prerequisite to Effective Modeling. Front Immunol 2019; 10:2522. [PMID: 31749803 PMCID: PMC6848063 DOI: 10.3389/fimmu.2019.02522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Most mathematical models that describe the individual or collective actions of cells aim at creating faithful representations of limited sets of data in a self-consistent manner. Consistency with relevant physiological rules pertaining to the greater picture is rarely imposed. By themselves, such models have limited predictive or even explanatory value, contrary to standard claims. Here I try to show that a more critical examination of currently held paradigms is necessary and could potentially lead to models that pass the test of time. In considering the evolution of paradigms over the past decades I focus on the “smart surveillance” theory of how T cells can respond differentially, individually and collectively, to both self- and foreign antigens depending on various “contextual” parameters. The overall perspective is that physiological messages to cells are encoded not only in the biochemical connections of signaling molecules to the cellular machinery but also in the magnitude, kinetics, and in the time- and space-contingencies, of sets of stimuli. By rationalizing the feasibility of subthreshold interactions, the “dynamic tuning hypothesis,” a central component of the theory, set the ground for further theoretical and experimental explorations of dynamically regulated immune tolerance, homeostasis and diversity, and of the notion that lymphocytes participate in nonclassical physiological functions. Some of these efforts are reviewed. Another focus of this review is the concomitant regulation of immune activation and homeostasis through the operation of a feedback mechanism controlling the balance between renewal and differentiation of activated cells. Different perspectives on the nature and regulation of chronic immune activation in HIV infection have led to conflicting models of HIV pathogenesis—a major area of research for theoretical immunologists over almost three decades—and can have profound impact on ongoing HIV cure strategies. Altogether, this critical review is intended to constructively influence the outlook of prospective model builders and of interested immunologists on the state of the art and to encourage conceptual work.
Collapse
Affiliation(s)
- Zvi Grossman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, United States.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
41
|
Huang HJ, Tsai YL, Lin SH, Hsu SH. Smart polymers for cell therapy and precision medicine. J Biomed Sci 2019; 26:73. [PMID: 31623607 PMCID: PMC6798433 DOI: 10.1186/s12929-019-0571-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/01/2019] [Indexed: 12/28/2022] Open
Abstract
Soft materials have been developed very rapidly in the biomedical field over the past 10 years because of advances in medical devices, cell therapy, and 3D printing for precision medicine. Smart polymers are one category of soft materials that respond to environmental changes. One typical example is the thermally-responsive polymers, which are widely used as cell carriers and in 3D printing. Self-healing polymers are one type of smart polymers that have the capacity to recover the structure after repeated damages and are often injectable through needles. Shape memory polymers are another type with the ability to memorize their original shape. These smart polymers can be used as cell/drug/protein carriers. Their injectability and shape memory performance allow them to be applied in bioprinting, minimally invasive surgery, and precision medicine. This review will describe the general materials design, characterization, as well as the current progresses and challenges of these smart polymers.
Collapse
Affiliation(s)
- Hung-Jin Huang
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan, Republic of China
| | - Yu-Liang Tsai
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan, Republic of China
| | - Shih-Ho Lin
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan, Republic of China
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan, Republic of China.
- Research and Development Center for Medical Devices, National Taiwan University, Taipei, Taiwan.
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35 Keyan Road, Miaoli, 35053, Taiwan, Republic of China.
| |
Collapse
|
42
|
Bourzac C, Bensidhoum M, Pallu S, Portier H. Use of adult mesenchymal stromal cells in tissue repair: impact of physical exercise. Am J Physiol Cell Physiol 2019; 317:C642-C654. [PMID: 31241985 PMCID: PMC6850997 DOI: 10.1152/ajpcell.00530.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 02/06/2023]
Abstract
Physical exercise (PE) has unquestionable beneficial effects on health, which likely extend into several organ-to-cell physiological processes. At the cell scale, endogenous mesenchymal stromal cells (MSCs) contribute to tissue repair, although their repair capacities may be insufficient in paucicellular or severely damaged tissues. For this reason, MSC transplantation holds great promise for tissue repair. With the goals of understanding if PE has beneficial effects on MSC biology and if PE potentiates their role in tissue repair, we reviewed literature reports regarding the effects of PE on MSC properties (specifically, proliferation, differentiation, and homing) and of a combination of PE and MSC transplantation on tissue repair (specifically neural, cartilage, and muscular tissues). Contradictory results have been reported; interpretation is complicated because various and different species, cell sources, and experimental protocols, specifically exercise programs, have been used. On the basis of these data, the effects of exercise on MSC proliferation and differentiation depend on exercise characteristics (type, intensity, duration, etc.) and on the characteristics of the tissue from which the MSCs were collected. For the in vitro studies, the level of strain (and other details of the mechanical stimulus), the time elapsed between the end of exposure to strain and MSC collection, the age of the donors, as well as the passage number at which the MSCs are evaluated also play a role. The combination of PE and MSC engraftment improves neural, cartilage, and muscular tissue recovery, but it is not clear whether the effects of MSCs and exercise are additive or synergistic.
Collapse
Affiliation(s)
- Celine Bourzac
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Ecole Nationale Vétérinaire d'Alfort, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Maisons-Alfort, France
| | - Morad Bensidhoum
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
| | - Stephane Pallu
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| | - Hugues Portier
- Université de Paris, CNRS, INSERM, Laboratoire de Biologie, Bioingenierie et Bioimagerie Osteoarticulaires (B3OA), Paris, France
- Université d'Orléans, Le Collegium sciences et techniques (COST), Orléans, France
| |
Collapse
|
43
|
Diaz-Navarro R, Urrútia G, Cleland JGF, Poloni D, Villagran F, Bangdiwala S, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Hippokratia 2019. [DOI: 10.1002/14651858.cd013433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Rienzi Diaz-Navarro
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Gerard Urrútia
- CIBER Epidemiología y Salud Pública (CIBERESP); Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau); Sant Antoni Maria Claret, 167 Pavilion 18 (D-53) Barcelona Catalonia Spain 08025
| | - John GF Cleland
- Imperial College London; National Heart and Lung Institute; London UK
| | - Daniel Poloni
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Francisco Villagran
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Shrikant Bangdiwala
- Collaborative Studies Coordinating Center; Department of Biostatistics, Gillings School of Global Public Health; Suite 203, Bank of America Center 137 E. Franklin Street Chapel Hill North Carolina USA 27514-4145
| | - Gabriel Rada
- Pontificia Universidad Católica de Chile; Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine; Lira 44, Decanato Primer piso Santiago Chile
| | - Eva Madrid
- Cochrane Centre School of Medicine Universidad de Valparaiso; Interdisciplinary Centre for Health Studies CIESAL; Viña del Mar Chile
- Universidad de Valparaiso; Chilean Cochrane Centre; Valparaiso Chile
| |
Collapse
|
44
|
Jang HJ, Lee EC, Kwon GJ, Seo YK. The effect of coated nano-hydroxyapatite concentration on scaffolds for osteogenesis. J Biomater Appl 2019; 34:827-839. [PMID: 31526073 DOI: 10.1177/0885328219875275] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hyun Jun Jang
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University Biomedi Campus, Gyeonggi-do, Korea
| | - Eun Cheol Lee
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University Biomedi Campus, Gyeonggi-do, Korea
| | | | - Young Kwon Seo
- Department of Medical Biotechnology (BK21 Plus team), Dongguk University Biomedi Campus, Gyeonggi-do, Korea
| |
Collapse
|
45
|
Pelissari C, Paris AFC, Mantesso A, Trierveiler M. Apical Papilla Cells Are Capable of Forming a Pulplike Tissue with Odontoblastlike Cells without the Use of Exogenous Growth Factors. J Endod 2019; 44:1671-1676. [PMID: 30409448 DOI: 10.1016/j.joen.2018.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/04/2018] [Accepted: 08/12/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Dental pulp is a complex tissue with highly differentiated cells, which makes its reconstruction a challenging task. The apical papilla is an undifferentiated tissue considered as the remnant of the dental papilla that forms the dentin-pulp complex. Aiming to analyze morphologic features of the tissue formed in an in vivo pulp model, we used human apical papilla as a cell source without the use of exogenous growth factors. METHODS A construct was built using newborn mice molar crowns treated with TrypLE (Fisher Scientific, Loughborough, UK) and EDTA. The crowns were filled with PuraMatrix (Corning Inc, Corning, NY) and a pool population of human apical papilla cells. As a control, we used crowns filled only with PuraMatrix and empty crowns. The constructs were transplanted under severe combined immunodeficient mice kidney capsules. Immunohistochemistry for lamin A, dentin sialophosphoprotein, and dentin matrix protein 1 was performed. RESULTS Morphologic analysis of all transplanted crowns showed the formation of a loose connective tissue of variable cellularity with the presence of well-formed functional vessels. In the study group, lamin A-positive cells represented the majority of cells within the pulp chamber and a few cells in the vessel lining. We also found positivity for dentin sialophosphoprotein and dentin matrix protein 1, an indicator of odontoblast differentiation. CONCLUSIONS In our study model, human transplanted apical papilla cells mixed with the host cells and formed a vascularized viable tissue, and these cells were able to differentiate into odontoblastlike cells without the use of exogenous growth factors.
Collapse
Affiliation(s)
- Cibele Pelissari
- Laboratory of Stem Cell Biology in Dentistry-LABITRON, Oral and Maxillofacial Pathology Department, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Adriana F C Paris
- Laboratory of Stem Cell Biology in Dentistry-LABITRON, Oral and Maxillofacial Pathology Department, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Andrea Mantesso
- Laboratory of Stem Cell Biology in Dentistry-LABITRON, Oral and Maxillofacial Pathology Department, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marília Trierveiler
- Laboratory of Stem Cell Biology in Dentistry-LABITRON, Oral and Maxillofacial Pathology Department, School of Dentistry, University of São Paulo, São Paulo, São Paulo, Brazil.
| |
Collapse
|
46
|
Hawsawi YM, Al-Zahrani F, Mavromatis CH, Baghdadi MA, Saggu S, Oyouni AAA. Stem Cell Applications for Treatment of Cancer and Autoimmune Diseases: Its Promises, Obstacles, and Future Perspectives. Technol Cancer Res Treat 2019; 17:1533033818806910. [PMID: 30343639 PMCID: PMC6198389 DOI: 10.1177/1533033818806910] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Since the original discovery of stem cells, a new era of promising results has emerged in the clinical application of stem cells for the treatment of several important diseases, including cancer and autoimmune diseases. The plentiful research on stem cells during the past decades has provided significant information on the developmental, morphological, and physiological processes that govern tissue and organ formation, maintenance, and regeneration; cellular differentiation; molecular processes; and tissue homeostasis. In this review, we present the history of the use of stem cells in different clinical applications. Furthermore, we discuss the various therapeutic options for stem cells in cancer, followed by the role of stem cells in the treatment of autoimmune disorders. Additionally, we highlight the risks of and obstacles to the application of stem cells in clinical practice. Ultimately, we show future perspectives in stem cell use, with an aim to improve the clinical usefulness of stem cells.
Collapse
Affiliation(s)
- Yousef M Hawsawi
- 1 Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.,2 Department of Biological Sciences, Faculty of Science and Arts, King Abdulaziz University, Rabigh, Kingdom of Saudi Arabia.,3 Department of Epidemiology and Biostatistics, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
| | - Faisal Al-Zahrani
- 2 Department of Biological Sciences, Faculty of Science and Arts, King Abdulaziz University, Rabigh, Kingdom of Saudi Arabia
| | - Charalampos Harris Mavromatis
- 2 Department of Biological Sciences, Faculty of Science and Arts, King Abdulaziz University, Rabigh, Kingdom of Saudi Arabia
| | - Mohammed A Baghdadi
- 1 Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia.,3 Department of Epidemiology and Biostatistics, King Faisal Specialist Hospital and Research Center, Jeddah, Kingdom of Saudi Arabia
| | - Shalini Saggu
- 4 Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Atif Abdulwahab A Oyouni
- 4 Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| |
Collapse
|
47
|
Afenya EK, Ouifki R, Mundle SD. Mathematical modeling of bone marrow - peripheral blood dynamics in the disease state based on current emerging paradigms, part II. J Theor Biol 2019; 460:37-55. [PMID: 30296448 DOI: 10.1016/j.jtbi.2018.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/28/2018] [Accepted: 10/01/2018] [Indexed: 12/31/2022]
Abstract
The cancer stem cell hypothesis has gained currency in recent times but concerns remain about its scientific foundations because of significant gaps that exist between research findings and comprehensive knowledge about cancer stem cells (CSCs). In this light, a mathematical model that considers hematopoietic dynamics in the diseased state of the bone marrow and peripheral blood is proposed and used to address findings about CSCs. The ensuing model, resulting from a modification and refinement of a recent model, develops out of the position that mathematical models of CSC development, that are few at this time, are needed to provide insightful underpinnings for biomedical findings about CSCs as the CSC idea gains traction. Accordingly, the mathematical challenges brought on by the model that mirror general challenges in dealing with nonlinear phenomena are discussed and placed in context. The proposed model describes the logical occurrence of discrete time delays, that by themselves present mathematical challenges, in the evolving cell populations under consideration. Under the challenging circumstances, the steady state properties of the model system of delay differential equations are obtained, analyzed, and the resulting mathematical predictions arising therefrom are interpreted and placed within the framework of findings regarding CSCs. Simulations of the model are carried out by considering various parameter scenarios that reflect different experimental situations involving disease evolution in human hosts. Model analyses and simulations suggest that the emergence of the cancer stem cell population alongside other malignant cells engenders higher dimensions of complexity in the evolution of malignancy in the bone marrow and peripheral blood at the expense of healthy hematopoietic development. The model predicts the evolution of an aberrant environment in which the malignant population particularly in the bone marrow shows tendencies of reaching an uncontrollable equilibrium state. Essentially, the model shows that a structural relationship exists between CSCs and non-stem malignant cells that confers on CSCs the role of temporally enhancing and stimulating the expansion of non-stem malignant cells while also benefitting from increases in their own population and these CSCs may be the main protagonists that drive the ultimate evolution of the uncontrollable equilibrium state of such malignant cells and these may have implications for treatment.
Collapse
Affiliation(s)
- Evans K Afenya
- Department of Mathematics, Elmhurst College, 190 Prospect Avenue, Elmhurst, IL 60126, USA.
| | - Rachid Ouifki
- Department of Mathematics and Applied Mathematics, University of Pretoria, South Africa.
| | - Suneel D Mundle
- Department of Biochemistry, Rush University Medical Center, 1735 W. Harrison St, Chicago, IL 60612, USA.
| |
Collapse
|
48
|
Aydin S, Şahin F. Stem Cells Derived from Dental Tissues. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1144:123-132. [DOI: 10.1007/5584_2018_333] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Abdik H, Avşar Abdik E, Demirci S, Doğan A, Turan D, Şahin F. The effects of bisphosphonates on osteonecrosis of jaw bone: a stem cell perspective. Mol Biol Rep 2018; 46:763-776. [PMID: 30506511 DOI: 10.1007/s11033-018-4532-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 11/28/2018] [Indexed: 12/29/2022]
Abstract
Bisphosphonate-induced osteonecrosis of the jaw (BIONJ) is a commonly encountered side effect of Bisphosphonates (BPs). Although certain aspects of BIONJ have been studied, the effects of BPs on the proliferation, differentiation, and maintenance of dental stem cells (DSC) in way that might account for development of BIONJ have not been evaluated. In the current study, Dental Pulp Stem Cells (DPSCs), Periodontal Stem Cells (PDLSCs), and human Tooth Germ Stem Cells (hTGSCs) were characterized and then each stem cell type were treated with selected BPs: Zoledronate (ZOL), Alendronate (ALE), and Risedronate (RIS). Negative effect on osteogenesis capacity of DSCs has not been observed after differentiation experiments in vitro. BPs exerted inhibitory effect on the migratory capacities of stem cells confirmed by in vitro scratch assay analysis. Angiogenesis of endothelial cells was blocked by BPs treatment in tube formation analysis. In conclusion, inhibitory effects of BPs on migration capacity of DSCs localized in close proximity to the jaw bone might be the primary reason for the side effects of BPs in the development of BIONJ process. Therefore, further in vivo evidence is required to investigate DSC properties in BP treated animals which might elucidate the importance of DSCs in BIONJ formation.
Collapse
Affiliation(s)
- Hüseyin Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Ezgi Avşar Abdik
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Selami Demirci
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, MD, USA
| | - Ayşegül Doğan
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Duygu Turan
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Fikrettin Şahin
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
50
|
Side Population: Its Use in the Study of Cellular Heterogeneity and as a Potential Enrichment Tool for Rare Cell Populations. Stem Cells Int 2018; 2018:2472137. [PMID: 30627171 PMCID: PMC6304857 DOI: 10.1155/2018/2472137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/07/2018] [Accepted: 11/14/2018] [Indexed: 12/16/2022] Open
Abstract
There is still much to learn about the cells used for cell- and gene-based therapies in the clinical setting. Stem cells are found in virtually all tissues in the human body. As a result, cells isolated from these tissues are a heterogeneous population consisting of various subpopulations including stem cells. Several strategies have been used to isolate and define the subpopulations that constitute these heterogeneous populations, one of which is the side population (SP) assay. SP cells are identified by their ability to efflux a fluorescent dye at a rate that is greater than the main cell population. This elevated rate of dye efflux has been attributed to the expression of members of the ATP-binding cassette (ABC) transporter protein family. SP cells have been identified in various tissues. In this review, we discuss the research to date on SP cells, focussing on SP cells identified in haematopoietic stem cells, adipose-derived stromal cells, and dental pulp.
Collapse
|