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
|
Sababathy M, Ramanathan G, Ganesan S, Sababathy S, Yasmin A, Ramasamy R, Foo J, Looi Q, Nur-Fazila S. Multipotent mesenchymal stromal/stem cell-based therapies for acute respiratory distress syndrome: current progress, challenges, and future frontiers. Braz J Med Biol Res 2024; 57:e13219. [PMID: 39417447 PMCID: PMC11484355 DOI: 10.1590/1414-431x2024e13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical, life-threatening condition marked by severe inflammation and impaired lung function. Mesenchymal stromal/stem cells (MSCs) present a promising therapeutic avenue due to their immunomodulatory, anti-inflammatory, and regenerative capabilities. This review comprehensively evaluates MSC-based strategies for ARDS treatment, including direct administration, tissue engineering, extracellular vesicles (EVs), nanoparticles, natural products, artificial intelligence (AI), gene modification, and MSC preconditioning. Direct MSC administration has demonstrated therapeutic potential but necessitates optimization to overcome challenges related to effective cell delivery, homing, and integration into damaged lung tissue. Tissue engineering methods, such as 3D-printed scaffolds and MSC sheets, enhance MSC survival and functionality within lung tissue. EVs and MSC-derived nanoparticles offer scalable and safer alternatives to cell-based therapies. Likewise, natural products and bioactive compounds derived from plants can augment MSC function and resilience, offering complementary strategies to enhance therapeutic outcomes. In addition, AI technologies could aid in optimizing MSC delivery and dosing, and gene editing tools like CRISPR/Cas9 allow precise modification of MSCs to enhance their therapeutic properties and target specific ARDS mechanisms. Preconditioning MSCs with hypoxia, growth factors, or pharmacological agents further enhances their therapeutic potential. While MSC therapies hold significant promise for ARDS, extensive research and clinical trials are essential to determine optimal protocols and ensure long-term safety and effectiveness.
Collapse
Affiliation(s)
- M. Sababathy
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - G. Ramanathan
- Faculty of Computer Science and Information Technology, University Malaya, Kuala Lumpur, Malaysia
| | - S. Ganesan
- School of Pharmacy, Management and Science University, Shah Alam, Selangor, Malaysia
| | - S. Sababathy
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Sungai Besi, Kuala Lumpur, Malaysia
| | - A.R. Yasmin
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - R. Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - J.B. Foo
- Center for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Q.H. Looi
- My Cytohealth Sdn. Bhd., Bandar Seri Petaling, Kuala Lumpur, Malaysia
| | - S.H. Nur-Fazila
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
3
|
Arévalo-Romero JA, López-Cantillo G, Moreno-Jiménez S, Marcos-Alcalde Í, Ros-Pardo D, Camacho BA, Gómez-Puertas P, Ramírez-Segura CA. In Silico Design of miniACE2 Decoys with In Vitro Enhanced Neutralization Activity against SARS-CoV-2, Encompassing Omicron Subvariants. Int J Mol Sci 2024; 25:10802. [PMID: 39409131 PMCID: PMC11476394 DOI: 10.3390/ijms251910802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
The COVID-19 pandemic has overwhelmed healthcare systems and triggered global economic downturns. While vaccines have reduced the lethality rate of SARS-CoV-2 to 0.9% as of October 2024, the continuous evolution of variants remains a significant public health challenge. Next-generation medical therapies offer hope in addressing this threat, especially for immunocompromised individuals who experience prolonged infections and severe illnesses, contributing to viral evolution. These cases increase the risk of new variants emerging. This study explores miniACE2 decoys as a novel strategy to counteract SARS-CoV-2 variants. Using in silico design and molecular dynamics, blocking proteins (BPs) were developed with stronger binding affinity for the receptor-binding domain of multiple variants than naturally soluble human ACE2. The BPs were expressed in E. coli and tested in vitro, showing promising neutralizing effects. Notably, miniACE2 BP9 exhibited an average IC50 of 4.9 µg/mL across several variants, including the Wuhan strain, Mu, Omicron BA.1, and BA.2 This low IC50 demonstrates the potent neutralizing ability of BP9, indicating its efficacy at low concentrations.Based on these findings, BP9 has emerged as a promising therapeutic candidate for combating SARS-CoV-2 and its evolving variants, thereby positioning it as a potential emergency biopharmaceutical.
Collapse
Affiliation(s)
- Jenny Andrea Arévalo-Romero
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Gina López-Cantillo
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Sara Moreno-Jiménez
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Íñigo Marcos-Alcalde
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - David Ros-Pardo
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - Bernardo Armando Camacho
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| | - Paulino Gómez-Puertas
- Grupo de Modelado Molecular del Centro de Biología Molecular Severo Ochoa, 14 CSIC-UAM, 28049 Madrid, Spain; (Í.M.-A.); (D.R.-P.)
| | - Cesar A. Ramírez-Segura
- Unidad de Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, Bogotá 111611, Colombia; (J.A.A.-R.); (G.L.-C.); (S.M.-J.); (B.A.C.)
| |
Collapse
|
4
|
Tang C, Dziedzic A, Khatib MN, Alhumaid S, Thangavelu L, Parameswari RP, Satapathy P, Zahiruddin QS, Rustagi S, Alanazi MA, Al-Thaqafy MS, Hazazi A, Alotaibi J, Al Faraj NJ, Al-Zaki NA, Al Marshood MJ, Al Saffar TY, Alsultan KA, Al-Ahmed SH, Rabaan AA. Stem cell therapy for COVID-19 treatment: an umbrella review. Int J Surg 2024; 110:6402-6417. [PMID: 38967503 PMCID: PMC11487013 DOI: 10.1097/js9.0000000000001786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/29/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND COVID-19 has presented significant obstacles to healthcare. Stem cell therapy, particularly mesenchymal stem cells, has emerged as a potential treatment modality due to its immunomodulatory and regenerative properties. This umbrella review aims to synthesize current evidence from systematic reviews on the safety and efficacy of stem cell therapy in COVID-19 treatment. METHODS A thorough literature search was performed across Embase, PubMed, Cochrane, and Web of Science from December 2019 to February 2024. Systematic reviews focusing on the use of stem cell therapy for COVID-19 were included. Evidence was synthesized by meta-analysis using R software (V 4.3) for each outcome. The certainty of evidence was assessed using the GRADE approach. RESULTS A total of 24 systematic reviews were included. Stem cell therapy was associated with reduced mortality [risk ratio (RR) 0.72, 95% CI: 0.60-0.86]; shorter hospital stays (mean difference -4.00 days, 95% CI: -4.68 to -3.32), and decreased need for invasive ventilation (RR 0.521, 95% CI: 0.320-0.847). Symptom remission rates improved (RR 1.151, 95% CI: 0.998-1.330), and a reduction in C-reactive protein levels was noted (standardized mean difference -1.198, 95% CI: -2.591 to 0.195), albeit with high heterogeneity. For adverse events, no significant differences were found between stem cell therapy and standard care (RR 0.87, 95% CI: 0.607-1.265). The certainty of evidence ranged from low to moderate. CONCLUSION Stem cell therapy demonstrates a potential benefit in treating COVID-19, particularly in reducing mortality and hospital stay duration. Despite these promising findings, the evidence is varied, and future large-scale randomized trials are essential to confirm the efficacy and optimize the therapeutic protocols for stem cell therapy in the management of the disease. The safety profile is encouraging, with no significant increase in adverse events, suggesting a viable avenue for treatment expansion.
Collapse
Affiliation(s)
- Chaozhi Tang
- College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Arkadiusz Dziedzic
- Department of Conservative Dentistry with Endodontics, Medical University of Silesia, Katowice, Poland
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education
| | - Saad Alhumaid
- School of Pharmacy, University of Tasmania, Hobart, Australia
| | - Lakshmi Thangavelu
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai
| | - RP Parameswari
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai
| | - Prakasini Satapathy
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Quazi Syed Zahiruddin
- South Asia Infant Feeding Research Network (SAIFRN), Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | | | - Majid S. Al-Thaqafy
- Infection Prevention and Control Department, King Abdulaziz Medical City, National Guard Health Affairs
- Epidemiology and Public Health, King Abdullah International Medical Research Center, National Guard Health Affairs
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, National Guard Health Affairs, Jeddah
| | - Ali Hazazi
- Department of Pathology and Laboratory Medicine, Security Forces Hospital Program, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Jawaher Alotaibi
- Infectious Diseases Unit, Department of Medicine, King Faisal Specialist Hospital and Research Center, Riyadh
| | | | | | | | | | | | | | - Ali A. Rabaan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| |
Collapse
|
5
|
Pouwels SD, Ter Haar EAMD, Heijink IH, Hylkema MN, Koster TD, Kuks PJM, Maassen S, Slebos DJ, Vasse GF, de Vries M, Woldhuis RR, Brandsma CA. Highlights from the 11th Bronchitis International Symposium: "Heterogeneity of Lung Disease in a Changing Environment," Groningen, The Netherlands, 2024. Respiration 2024; 103:765-776. [PMID: 39348815 DOI: 10.1159/000541655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024] Open
Abstract
This meeting report provides an overview of the highlights of the Bronchitis XI international symposium, held in June 2024 in Groningen, The Netherlands. The theme of this year's symposium was "heterogeneity of lung disease in a changing environment," and the symposium contained five different sessions focused on (i) heterogeneity of chronic lung disease, (ii) environmental changes with impact on lung disease, (iii) the aging lung, (iv) bronchitis, and (v) innovative therapy. The highlights from each of these sessions will be discussed separately, providing an overview of latest studies, new data, and enthralling discussions.
Collapse
Affiliation(s)
- Simon D Pouwels
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Else A M D Ter Haar
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Machteld N Hylkema
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - T David Koster
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Pauline J M Kuks
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Sjors Maassen
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Dirk-Jan Slebos
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pulmonary Diseases, University of Groningen, Groningen, The Netherlands
| | - Gwenda F Vasse
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - Maaike de Vries
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Epidemiology, University of Groningen, Groningen, The Netherlands
| | - Roy R Woldhuis
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
- University Medical Center Groningen, Department of Pathology and Medical Biology, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Chacon Alberty L, King M, Mesquita FCP, Hochman-Mendez C. Quality Assessment of Long-Term Cryopreserved Human Bone-Derived Marrow Mesenchymal Stromal Cell Samples: Experience from the Texas Heart Institute Biorepository and Biospecimen Profiling Core. Biopreserv Biobank 2024. [PMID: 39253842 DOI: 10.1089/bio.2023.0144] [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: 09/11/2024] Open
Abstract
In biomedical research, biorepositories are pivotal resources that safeguard and supply clinical samples for scientific investigators. Proper long-term cryopreservation conditions are essential to maintain biospecimen quality. In this study, we analyzed the efficacy of sample cryopreservation at the Texas Heart Institute Biorepository and Biospecimen Profiling Core (THI-BRC). Our assessments included a thorough review of internal processes, quality reports, and both internal and external audit outcomes. We examined the integrity of human bone marrow-derived multipotent mesenchymal stromal cells (BM-MSCs) that were cryopreserved for over 5 years. These samples originated from randomly selected clinical trial participants or commercially sourced cell lines. Parameters such as cell viability, DNA and RNA integrity, population doubling time, sterility, and BM-MSC-specific attributes such as surface antigen expression and differentiation potential were studied. BM-MSC samples cryopreserved for ∼6 months served as our control. Our results demonstrated that the 5-year cryopreserved samples maintained their integrity compared with the shorter-term stored control samples. Moreover, THI-BRC has met accreditation agency standards and has not received any repeated deficiencies over 7 years. Collectively, our findings affirm that THI-BRC's biospecimen storage protocols align with accepted standards as confirmed by the quality assessment of long-term stored clinical samples.
Collapse
Affiliation(s)
- Lourdes Chacon Alberty
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
- Regenerative Medicine Research, The Texas Heart Institute, Houston, Texas, USA
| | - Madelyn King
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
| | | | - Camila Hochman-Mendez
- Biorepository and Biospecimen Profiling Core, The Texas Heart Institute, Houston, Texas, USA
- Regenerative Medicine Research, The Texas Heart Institute, Houston, Texas, USA
| |
Collapse
|
7
|
Zheng Q, Li Y, Sheng G, Li L. The Value of Ursodeoxycholic Acid and Mesenchymal Stem Cells in the Treatment of Severe COVID-19. Microorganisms 2024; 12:1269. [PMID: 39065038 PMCID: PMC11279161 DOI: 10.3390/microorganisms12071269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Objective: The objective of this study was to evaluate the therapeutic efficacy of ursodeoxycholic acid (UDCA) and mesenchymal stem cells (MSCs) in patients with severe COVID-19. Methods: We included severe COVID-19 patients hospitalized at Shulan (Hangzhou) Hospital between December 2022 and June 2023. We used a logistic regression model to compare the use of UDCA and MSCs in the two distinct groups of improved and poor outcomes. It is noteworthy that the deterioration group encompassed instances of both death and abandonment of treatment. The receiver operating characteristic (ROC) curve was plotted to assess the performance of the model. The aim was to assess the therapeutic effect of UDCA and MSCs on the outcome of severe COVID-19 patients. Results: A total of 167 patients with severe COVID-19 were included in this study. The analysis revealed that out of 42 patients (25.1%), 17 patients (10.2%) had taken UDCA, and 17 patients (10.2%) had used MSCs. Following a multivariable logistic regression, the results indicated a negative association between UDCA treatment (OR = 0.38 (0.16-0.91), p = 0.029), MSCs treatment (OR = 0.21 (0.07-0.65), p = 0.007), and the risk of severe COVID-19 mortality. Additionally, age showed a positive association with the risk of mortality (OR = 1.03 (1.01-1.07), p = 0.025). Conclusions: UDCA and MSCs have shown potential in improving the prognosis of severe COVID-19 patients and could be considered as additional treatments for COVID-19 in the future.
Collapse
Affiliation(s)
- Qi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| | - Yuetong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| | - Guoping Sheng
- Department of Infectious Disease, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou 310022, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China; (Q.Z.); (Y.L.)
| |
Collapse
|
8
|
Marquez-Curtis LA, Elliott JAW. Mesenchymal stromal cells derived from various tissues: Biological, clinical and cryopreservation aspects: Update from 2015 review. Cryobiology 2024; 115:104856. [PMID: 38340887 DOI: 10.1016/j.cryobiol.2024.104856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Mesenchymal stromal cells (MSCs) have become one of the most investigated and applied cells for cellular therapy and regenerative medicine. In this update of our review published in 2015, we show that studies continue to abound regarding the characterization of MSCs to distinguish them from other similar cell types, the discovery of new tissue sources of MSCs, and the confirmation of their properties and functions that render them suitable as a therapeutic. Because cryopreservation is widely recognized as the only technology that would enable the on-demand availability of MSCs, here we show that although the traditional method of cryopreserving cells by slow cooling in the presence of 10% dimethyl sulfoxide (Me2SO) continues to be used by many, several novel MSC cryopreservation approaches have emerged. As in our previous review, we conclude from these recent reports that viable and functional MSCs from diverse tissues can be recovered after cryopreservation using a variety of cryoprotectants, freezing protocols, storage temperatures, and periods of storage. We also show that for logistical reasons there are now more studies devoted to the cryopreservation of tissues from which MSCs are derived. A new topic included in this review covers the application in COVID-19 of MSCs arising from their immunomodulatory and antiviral properties. Due to the inherent heterogeneity in MSC populations from different sources there is still no standardized procedure for their isolation, identification, functional characterization, cryopreservation, and route of administration, and not likely to be a "one-size-fits-all" approach in their applications in cell-based therapy and regenerative medicine.
Collapse
Affiliation(s)
- Leah A Marquez-Curtis
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9
| | - Janet A W Elliott
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada, T6G 1H9; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada, T6G 1C9.
| |
Collapse
|
9
|
Martínez-Muñoz ME, Payares-Herrera C, Lipperheide I, Malo de Molina R, Salcedo I, Alonso R, Martín-Donaire T, Sánchez R, Zafra R, García-Berciano M, Trisán-Alonso A, Pérez-Torres M, Ramos-Martínez A, Ussetti P, Rubio JJ, Avendaño-Solà C, Duarte RF. Mesenchymal stromal cell therapy for COVID-19 acute respiratory distress syndrome: a double-blind randomised controlled trial. Bone Marrow Transplant 2024; 59:777-784. [PMID: 38409332 DOI: 10.1038/s41409-024-02230-5] [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: 07/14/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/28/2024]
Abstract
Mesenchymal stromal cells (MSC) have immunomodulatory and tissue-regenerative properties and have shown promising results in acute respiratory distress syndrome (ARDS) of multiple causes, including COVID-19. We conducted a randomised (1:1), placebo-controlled, double-blind clinical trial to assess the efficacy and safety of one bone marrow-derived MSC infusion in twenty patients with moderate to severe ARDS caused by COVID-19. The primary endpoint (increase in PaO2/FiO2 ratio from baseline to day 7, MSC 83.3 versus placebo 57.6) was not statistically significant, although a clinical improvement at day 7 in the WHO scale was observed in MSC patients (5, 50% vs 0, 0%, p = 0.033). Median time to discontinuation of supplemental oxygen was also shorter in the experimental arm (14 versus 23 days, p = 0.007), resulting in a shorter hospital stay (17.5 versus 28 days, p = 0.042). No significant differences were observed for other efficacy or safety secondary endpoints. No infusion or treatment-related serious adverse events occurred during the one-year follow-up. This study did not meet the primary endpoint of PaO2/FiO2 increase by day 7, although it suggests that MSC are safe in COVID-19 ARDS and may accelerate patients' clinical recovery and hospital discharge. Larger studies are warranted to elucidate their role in ARDS and other inflammatory lung disorders.Trial Registration: EudraCT Number: 2020-002193-27, registered on July 14th, 2020, https://www.clinicaltrialsregister.eu/ctr-search/trial/2020-002193-27/ES . NCT number: NCT04615429, registered on November 4th, 2020, https://clinicaltrials.gov/ct2/show/NCT04615429 .
Collapse
Affiliation(s)
- María E Martínez-Muñoz
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Concepción Payares-Herrera
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Inés Lipperheide
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Rosa Malo de Molina
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Isabel Salcedo
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rosalía Alonso
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Trinidad Martín-Donaire
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rocío Sánchez
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Rocío Zafra
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Miguel García-Berciano
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
| | - Andrea Trisán-Alonso
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Manuel Pérez-Torres
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Antonio Ramos-Martínez
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Internal Medicine and Infectious Diseases, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Piedad Ussetti
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Pneumology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Juan J Rubio
- Intensive Care Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Cristina Avendaño-Solà
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain
- Department of Clinical Pharmacology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Rafael F Duarte
- Department of Haematology and GMP Cellular Therapy Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain.
- Instituto de Investigación Sanitaria Puerta de Hierro Segovia Arana, Madrid, Spain.
| |
Collapse
|
10
|
Arevalo-Romero JA, Chingaté-López SM, Camacho BA, Alméciga-Díaz CJ, Ramirez-Segura CA. Next-generation treatments: Immunotherapy and advanced therapies for COVID-19. Heliyon 2024; 10:e26423. [PMID: 38434363 PMCID: PMC10907543 DOI: 10.1016/j.heliyon.2024.e26423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 03/05/2024] Open
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in 2019 following prior outbreaks of coronaviruses like SARS and MERS in recent decades, underscoring their high potential of infectivity in humans. Insights from previous outbreaks of SARS and MERS have played a significant role in developing effective strategies to mitigate the global impact of SARS-CoV-2. As of January 7, 2024, there have been 774,075,242 confirmed cases of COVID-19 worldwide. To date, 13.59 billion vaccine doses have been administered, and there have been 7,012,986 documented fatalities (https://www.who.int/) Despite significant progress in addressing the COVID-19 pandemic, the rapid evolution of SARS-CoV-2 challenges human defenses, presenting ongoing global challenges. The emergence of new SARS-CoV-2 lineages, shaped by mutation and recombination processes, has led to successive waves of infections. This scenario reveals the need for next-generation vaccines as a crucial requirement for ensuring ongoing protection against SARS-CoV-2. This demand calls for formulations that trigger a robust adaptive immune response without leading the acute inflammation linked with the infection. Key mutations detected in the Spike protein, a critical target for neutralizing antibodies and vaccine design -specifically within the Receptor Binding Domain region of Omicron variant lineages (B.1.1.529), currently dominant worldwide, have intensified concerns due to their association with immunity evasion from prior vaccinations and infections. As the world deals with this evolving threat, the narrative extends to the realm of emerging variants, each displaying new mutations with implications that remain largely misunderstood. Notably, the JN.1 Omicron lineage is gaining global prevalence, and early findings suggest it stands among the immune-evading variants, a characteristic attributed to its mutation L455S. Moreover, the detrimental consequences of the novel emergence of SARS-CoV-2 lineages bear a particularly critical impact on immunocompromised individuals and older adults. Immunocompromised individuals face challenges such as suboptimal responses to COVID-19 vaccines, rendering them more susceptible to severe disease. Similarly, older adults have an increased risk of severe disease and the presence of comorbid conditions, find themselves at a heightened vulnerability to develop COVID-19 disease. Thus, recognizing these intricate factors is crucial for effectively tailoring public health strategies to protect these vulnerable populations. In this context, this review aims to describe, analyze, and discuss the current progress of the next-generation treatments encompassing immunotherapeutic approaches and advanced therapies emerging as complements that will offer solutions to counter the disadvantages of the existing options. Preliminary outcomes show that these strategies target the virus and address the immunomodulatory responses associated with COVID-19. Furthermore, the capacity to promote tissue repair has been demonstrated, which can be particularly noteworthy for immunocompromised individuals who stand as vulnerable actors in the global landscape of coronavirus infections. The emerging next-generation treatments possess broader potential, offering protection against a wide range of variants and enhancing the ability to counter the impact of the constant evolution of the virus. Furthermore, advanced therapies are projected as potential treatment alternatives for managing Chronic Post-COVID-19 syndromeand addressing its associated long-term complications.
Collapse
Affiliation(s)
- Jenny Andrea Arevalo-Romero
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Sandra M. Chingaté-López
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Bernardo Armando Camacho
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| | - Carlos Javier Alméciga-Díaz
- Instituto de Errores Innatos del Metabolismo, Facultad de Ciencias, Pontificia Universidad Javeriana, 110231, Bogotá, D.C., Colombia
| | - Cesar A. Ramirez-Segura
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia, Biotecnología e Innovación en Salud, IDCBIS, 111611, Bogotá, DC, Colombia
| |
Collapse
|
11
|
Nagamura-Inoue T, Nagamura F. Umbilical cord blood and cord tissue banking as somatic stem cell resources to support medical cell modalities. Inflamm Regen 2023; 43:59. [PMID: 38053217 PMCID: PMC10696687 DOI: 10.1186/s41232-023-00311-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/21/2023] [Indexed: 12/07/2023] Open
Abstract
Human umbilical cord blood (CB) and umbilical cord tissue (UC) are attractive sources of somatic stem cells for gene and cell therapies. CB and UC can be obtained noninvasively from donors. CB, a known source of hematopoietic stem cells for transplantation, has attracted attention as a new source of immune cells, including universal chimeric antigen receptor-T cell therapy (CAR-T) and, more recently, universal CAR-natural killer cells. UC-derived mesenchymal stromal cells (UC-MSCs) have a higher proliferation potency than those derived from adult tissues and can be used anon-HLA restrictively. UC-MSCs meet the MSC criteria outlined by the International Society of Gene and Cellular Therapy. UC-MSCs are negative for HLA-DR, CD80, and CD86 and have an immunosuppressive ability that mitigates the proliferation of activated lymphocytes through secreting indoleamine 2,3-dioxygenase 1 and prostaglandin E2, and the expression of PD-L2 and PD-L1. We established the off-the-shelf cord blood/cord bank IMSUT CORD to support novel cell therapy modalities, including the CB-derived immune cells, MSCs, MSCs-derived extracellular vesicles, biological carriers loaded with chemotherapy drugs, prodrug, oncolytic viruses, nanoparticles, human artificial chromosome, combinational products with a scaffold, bio3D printing, and so on.
Collapse
Affiliation(s)
- Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Research Hospital, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Fumitaka Nagamura
- IMSUT CORD, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Advanced Medicine Promotion, The Advanced Clinical Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Sababathy M, Ramanathan G, Abd Rahaman NY, Ramasamy R, Biau FJ, Qi Hao DL, Hamid NFS. A 'one stone, two birds' approach with mesenchymal stem cells for acute respiratory distress syndrome and Type II diabetes mellitus. Regen Med 2023; 18:913-934. [PMID: 38111999 DOI: 10.2217/rme-2023-0193] [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] [Indexed: 12/20/2023] Open
Abstract
This review explores the intricate relationship between acute respiratory distress syndrome (ARDS) and Type II diabetes mellitus (T2DM). It covers ARDS epidemiology, etiology and pathophysiology, along with current treatment trends and challenges. The lipopolysaccharides (LPS) role in ARDS and its association between non-communicable diseases and COVID-19 are discussed. The review highlights the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for ARDS and T2DM, emphasizing their immunomodulatory effects. This review also underlines how T2DM exacerbates ARDS pathophysiology and discusses the potential of hUC-MSCs in modulating immune responses. In conclusion, the review highlights the multidisciplinary approach to managing ARDS and T2DM, focusing on inflammation, oxidative stress and potential therapy of hUC-MSCs in the future.
Collapse
Affiliation(s)
- Mogesh Sababathy
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ghayathri Ramanathan
- Faculty of Computer Science & Information Technology, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Yasmin Abd Rahaman
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Foo Jhi Biau
- Centre for Drug Discovery & Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn. Bhd., 18-2, Jalan Radin Bagus 1, Bandar Seri Petaling, Kuala Lumpur, 57000, Malaysia
| | - Nur-Fazila Saulol Hamid
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
13
|
Chen X, Liu B, Li C, Wang Y, Geng S, Du X, Weng J, Lai P. Stem cell-based therapy for COVID-19. Int Immunopharmacol 2023; 124:110890. [PMID: 37688914 DOI: 10.1016/j.intimp.2023.110890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023]
Abstract
While The World Health Organization (WHO) has announced that COVID-19 is no longer a public health emergency of international concern(PHEIC), the risk of reinfection and new emerging variants still makes it crucial to study and work towards the prevention of COVID-19. Stem cell and stem cell-like derivatives have shown some promising results in clinical trials and preclinical studies as an alternative treatment option for the pulmonary illnesses caused by the COVID-19 and can be used as a potential vaccine. In this review, we will systematically summarize the pathophysiological process and potential mechanisms underlying stem cell-based therapy in COVID-19, and the registered COVID-19 clinical trials, and engineered extracellular vesicle as a potential vaccine for preventing COVID-19.
Collapse
Affiliation(s)
- Xiaomei Chen
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Bowen Liu
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Chao Li
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Yulian Wang
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Suxia Geng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, PR China.
| |
Collapse
|
14
|
Moll G, Luecht C, Gyamfi MA, da Fonseca DLM, Wang P, Zhao H, Gong Z, Chen L, Ashraf MI, Heidecke H, Hackel AM, Dragun D, Budde K, Penack O, Riemekasten G, Cabral-Marques O, Witowski J, Catar R. Autoantibodies from patients with kidney allograft vasculopathy stimulate a proinflammatory switch in endothelial cells and monocytes mediated via GPCR-directed PAR1-TNF-α signaling. Front Immunol 2023; 14:1289744. [PMID: 37965310 PMCID: PMC10642342 DOI: 10.3389/fimmu.2023.1289744] [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: 09/06/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Non-HLA-directed regulatory autoantibodies (RABs) are known to target G-protein coupled receptors (GPCRs) and thereby contribute to kidney transplant vasculopathy and failure. However, the detailed underlying signaling mechanisms in human microvascular endothelial cells (HMECs) and immune cells need to be clarified in more detail. In this study, we compared the immune stimulatory effects and concomitant intracellular and extracellular signaling mechanisms of immunoglobulin G (IgG)-fractions from kidney transplant patients with allograft vasculopathy (KTx-IgG), to that from patients without vasculopathy, or matched healthy controls (Con-IgG). We found that KTx-IgG from patients with vasculopathy, but not KTx-IgG from patients without vasculopathy or Con-IgG, elicits HMEC activation and subsequent upregulation and secretion of tumor necrosis factor alpha (TNF-α) from HMECs, which was amplified in the presence of the protease-activated thrombin receptor 1 (PAR1) activator thrombin, but could be omitted by selectively blocking the PAR1 receptor. The amount and activity of the TNF-α secreted by HMECs stimulated with KTx-IgG from patients with vasculopathy was sufficient to induce subsequent THP-1 monocytic cell activation. Furthermore, AP-1/c-FOS, was identified as crucial transcription factor complex controlling the KTx-IgG-induced endothelial TNF-α synthesis, and mircoRNA-let-7f-5p as a regulatory element in modulating the underlying signaling cascade. In conclusion, exposure of HMECs to KTx-IgG from patients with allograft vasculopathy, but not KTx-IgG from patients without vasculopathy or healthy Con-IgG, triggers signaling through the PAR1-AP-1/c-FOS-miRNA-let7-axis, to control TNF-α gene transcription and TNF-α-induced monocyte activation. These observations offer a greater mechanistic understanding of endothelial cells and subsequent immune cell activation in the clinical setting of transplant vasculopathy that can eventually lead to transplant failure, irrespective of alloantigen-directed responses.
Collapse
Affiliation(s)
- Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
- Berlin Institute of Healthy (BIH) Center for Regenerative Therapies (BCRT) and Berlin-Brandenburg School for Regenerative Therapies (BSRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Luecht
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Michael Adu Gyamfi
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Dennyson L M da Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, Brazil
| | - Pinchao Wang
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Hongfan Zhao
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Zexian Gong
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Lei Chen
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | | | | | | | - Duska Dragun
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| | - Olaf Penack
- Department of Hematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, Lübeck, Germany
| | - Otávio Cabral-Marques
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo (USP), São Paulo, Brazil
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, USP, São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, USP School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, USP School of Medicine, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, USP, São Paulo, Brazil
| | - Janusz Witowski
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
- Department of Pathophysiology, Poznan University of Medical Sciences, Poznan, Poland
| | - Rusan Catar
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Healthy (BIH), Berlin, Germany
| |
Collapse
|
15
|
Wang Y, Liang Q, Chen F, Zheng J, Chen Y, Chen Z, Li R, Li X. Immune-Cell-Based Therapy for COVID-19: Current Status. Viruses 2023; 15:2148. [PMID: 38005826 PMCID: PMC10674523 DOI: 10.3390/v15112148] [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: 08/19/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 11/26/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a global pandemic. The interplay between innate and adaptive immune responses plays a crucial role in managing COVID-19. Cell therapy has recently emerged as a promising strategy to modulate the immune system, offering immense potential for the treatment of COVID-19 due to its customizability and regenerative capabilities. This review provides an overview of the various subsets of immune cell subsets implicated in the pathogenesis of COVID-19 and a comprehensive summary of the current status of immune cell therapy in COVID-19 treatment.
Collapse
Affiliation(s)
- Yiyuan Wang
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qinghe Liang
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fengsheng Chen
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiehuang Zheng
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Chen
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ziye Chen
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruopeng Li
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaojuan Li
- Laboratory of Anti-Inflammatory and Immunomodulatory Pharmacology, Innovation Program of Drug Research on Inflammatory and Immune Diseases, Southern Medical University, Guangzhou 510515, China; (Y.W.); (Q.L.); (F.C.); (J.Z.); (Y.C.); (Z.C.); (R.L.)
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Herzig MC, Christy BA, Montgomery RK, Cantu-Garza C, Barrera GD, Lee JH, Mucha N, Talackine JR, Abaasah IA, Bynum JA, Cap AP. Short-term assays for mesenchymal stromal cell immunosuppression of T-lymphocytes. Front Immunol 2023; 14:1225047. [PMID: 37822938 PMCID: PMC10562633 DOI: 10.3389/fimmu.2023.1225047] [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: 05/18/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction Trauma patients are susceptible to coagulopathy and dysfunctional immune responses. Mesenchymal stromal cells (MSCs) are at the forefront of the cellular therapy revolution with profound immunomodulatory, regenerative, and therapeutic potential. Routine assays to assess immunomodulation activity examine MSC effects on proliferation of peripheral blood mononuclear cells (PBMCs) and take 3-7 days. Assays that could be done in a shorter period of time would be beneficial to allow more rapid comparison of different MSC donors. The studies presented here focused on assays for MSC suppression of mitogen-stimulated PBMC activation in time frames of 24 h or less. Methods Three potential assays were examined-assays of apoptosis focusing on caspase activation, assays of phosphatidyl serine externalization (PS+) on PBMCs, and measurement of tumor necrosis factor alpha (TNFα) levels using rapid ELISA methods. All assays used the same initial experimental conditions: cryopreserved PBMCs from 8 to 10 pooled donors, co-culture with and without MSCs in 96-well plates, and PBMC stimulation with mitogen for 2-72 h. Results Suppression of caspase activity in activated PBMCs by incubation with MSCs was not robust and was only significant at times after 24 h. Monitoring PS+ of live CD3+ or live CD4+/CD3+ mitogen-activated PBMCs was dose dependent, reproducible, robust, and evident at the earliest time point taken, 2 h, although no increase in the percentage of PS+ cells was seen with time. The ability of MSC in co-culture to suppress PBMC PS+ externalization compared favorably to two concomitant assays for MSC co-culture suppression of PBMC proliferation, at 72 h by ATP assay, or at 96 h by fluorescently labeled protein signal dilution. TNFα release by mitogen-activated PBMCs was dose dependent, reproducible, robust, and evident at the earliest time point taken, with accumulating signal over time. However, suppression levels with MSC co-culture was reliably seen only after 24 h. Discussion Takeaways from these studies are as follows: (1) while early measures of PBMC activation is evident at 2-6 h, immunosuppression was only reliably detected at 24 h; (2) PS externalization at 24 h is a surrogate assay for MSC immunomodulation; and (3) rapid ELISA assay detection of TNFα release by PBMCs is a robust and sensitive assay for MSC immunomodulation at 24 h.
Collapse
Affiliation(s)
- Maryanne C. Herzig
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Barbara A. Christy
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Robbie K. Montgomery
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Carolina Cantu-Garza
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Gema D. Barrera
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Ji H. Lee
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Nicholas Mucha
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Jennifer R. Talackine
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Isaac A. Abaasah
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - James A. Bynum
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
- Department of Surgery, University of Texas, Health Science Center, San Antonio, TX, United States
| | - Andrew P. Cap
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| |
Collapse
|