1
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [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: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Ivantcova PM, Sungatullina AR, Pidgirnaya KV, Nikitin MP. Exploring the synergy between bioluminescence and nanomaterials: Innovations in analytical and therapeutic applications. Colloids Surf B Biointerfaces 2025; 251:114631. [PMID: 40127545 DOI: 10.1016/j.colsurfb.2025.114631] [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: 09/30/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/26/2025]
Abstract
The application of bioluminescent luciferin-luciferase systems for visualizing and stimulating various processes in living systems is of great interest due to its specific nature and high signal-to-noise ratio. Nanomaterials can finely manipulate multiple parameters of the bioluminescent systems, including the enzyme stability, intensity, and duration of the irradiation. Also, bioluminescence can affect the properties of a nanomaterial, namely, to carry out BRET, to trigger cascades of various photochemical transformations. Here we summarize cases of the interplay between nanomaterials and various bioluminescent systems to improve various biosensors, biovisualization in cellulo, in vivo, and for therapy over the past twenty years. We reviewed interactions between a wide range of nanomaterials and bioluminescent systems, including bacterial and genetically encoded luciferases. This review aims to serve as a comprehensive guide for developing bioluminescent multimodal nanoplatforms for analytic applications and therapy.
Collapse
Affiliation(s)
- Polina M Ivantcova
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia.
| | - Adilya R Sungatullina
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia; Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russian Federation
| | - Kristina V Pidgirnaya
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia
| | - Maxim P Nikitin
- Sirius University of Science and Technology, 1 Olimpiyskiy Ave, 354340, Sirius, Krasnodar region, Russia; Moscow Center for Advanced Studies, Kulakova str. 20, Moscow 123592, Russian Federation
| |
Collapse
|
3
|
Martins F, Ribeiro MHL. Quality and Regulatory Requirements for the Manufacture of Master Cell Banks of Clinical Grade iPSCs: The EU and USA Perspectives. Stem Cell Rev Rep 2025; 21:645-679. [PMID: 39821060 DOI: 10.1007/s12015-024-10838-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) and protocols for their differentiation into various cell types have revolutionized the field of tissue engineering and regenerative medicine. Developing manufacturing guidelines for safe and GMP-compliant final products has become essential. Allogeneic iPSCs-derived cell therapies are now the preferred manufacturing alternative. This option requires the establishment of clinical-grade master cell banks of iPSCs. This study aimed at reviewing the Quality and Regulatory requirements from the two main authorities in the world-Europe (EMA) and the United States (FDA)-regarding the manufacture of clinical grade master cell banks (iPSCs). The minimum requirements for iPSCs to be used in first-in-human clinical trials were also reviewed, as well as current best practices currently followed by iPSC bank manufacturers for final product characterisation. The methodology used for this work was a review of various sources of information ranging from scientific literature, published guidance documents available on the EMA and FDA websites, GMP and ICH guidelines, and applicable compendial monographs. Manufacturers of iPSCs cell banks looking to qualify them for clinical use are turning to the ICH guidelines and trying to adapt their requirements. Specifically with the impact of the field of iPSC cell banks, the following areas should be subject to guidance and harmonisation: i) expression vectors authorized for iPSC generation; ii) minimum identity testing; iii) minimum purity testing (including adventitious agent testing); and iv) stability testing. Current ICH guidelines for biotechnological/biological products should be extended to cover cell banks used for cell therapies.
Collapse
Affiliation(s)
- Fernando Martins
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
- Stemamatters S.A., 4805-017, Guimarães, Portugal
| | - Maria H L Ribeiro
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
- Research Institute for Medicines (i-Med.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
| |
Collapse
|
4
|
Conte L, De Nunzio G, Lupo R, Cascio D, Cioce M, Vitale E, Ianne C, Rubbi I, Martino M, Lombardini L, Vassanelli A, Pupella S, Pollichieni S, Sacchi N, Ciceri F, Botti S. Raising awareness may increase the likelihood of hematopoietic stem cell donation: a nationwide survey using artificial intelligence. Int J Hematol 2025; 121:511-525. [PMID: 39668285 DOI: 10.1007/s12185-024-03894-x] [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: 07/11/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND In Italy, the demand for allogeneic transplantation exceeds the number of compatible donors in the Italian Bone Marrow Donor Registry (IBMDR). This study aimed to explore the knowledge, beliefs, opinions, values, and feelings of the Italian population regarding stem cell donation. METHODS An online survey was shared via social media. Respondents were retrospectively identified as registered on the IBMDR (donor group) or never registered (non-donor group). Statistical analyses confirmed the relationship between knowledge level and willingness to donate. Six machine learning classifiers were trained using questionnaire responses to predict the probability of IBMDR registration. RESULTS A total of 1518 respondents participated. Characteristics identified in the non-donor group were a lower level of knowledge regarding donation needs (51.7% vs 24.4%, p < 0.001) and negative feelings such as fear (Z = - 2.2642, p = 0.02), confusion (Z = 4.4821, p < 0.001), and uncertainty (Z = 3.3425, p < 0.001). Higher knowledge predicted a greater likelihood of IBMDR enrollment. Machine learning analysis showed an AUC ranging from 0.65 to 0.81, depending on the classifier. CONCLUSIONS The results underscore the need to improve strategies to raise awareness and knowledge of stem cell donation among the Italian population.
Collapse
Affiliation(s)
- Luana Conte
- Department of Physics and Chemistry, University of Palermo, Palermo, Italy
- Laboratory of Advanced Data Analysis for Medicine (ADAM), University of Salento, Lecce, Italy
| | - Giorgio De Nunzio
- Laboratory of Advanced Data Analysis for Medicine (ADAM), University of Salento, Lecce, Italy.
- Laboratory of Biomedical Physics and Environment, Department of Mathematics and Physics "E. De Giorgi", University of Salento, Lecce, Italy.
| | - Roberto Lupo
- San Giuseppe da Copertino Hospital, Local Health Authority, Copertino, Lecce, Italy
| | - Donato Cascio
- Department of Physics and Chemistry, University of Palermo, Palermo, Italy
| | - Marco Cioce
- Department UOC SITRA, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Chiara Ianne
- Comunità Riabilitativa Assistenziale Psichiatrica, "Carrubo", Avetrana, Taranto, Italy
| | - Ivan Rubbi
- School of Nursing, University of Bologna, Bologna, Italy
| | - Massimo Martino
- Grande Ospedale Metropolitano Bianchi Melacrino Morelli, Centro Unico Trapianti A. Neri, Reggio Calabria, Italy
| | | | - Aurora Vassanelli
- Transfusion Medicine Department, University and Hospital Trust (AOUI), Verona, Italy
| | | | | | - Nicoletta Sacchi
- Hematology and Hematopoietic Stem Cell Transplantation Unit, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- SOC Ematologia, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefano Botti
- Hematology and Hematopoietic Stem Cell Transplantation Unit, Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
5
|
Sharf-Pauker N, Galil I, Kfir O, Chen G, Menachem R, Shklover J, Schroeder A, Ackerman S. Scaling Up Synthetic Cell Production Using Robotics and Machine Learning Toward Therapeutic Applications. Adv Biol (Weinh) 2025:e2400671. [PMID: 40162738 DOI: 10.1002/adbi.202400671] [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: 10/31/2024] [Revised: 02/23/2025] [Indexed: 04/02/2025]
Abstract
Synthetic cells (SCs), developed through bottom-up synthetic biology, hold great potential for biomedical applications, with the promise of replacing malfunctioning natural cells and treating diseases with spatiotemporal control. Currently, most SC synthesis and characterization processes are manual, limiting scalability and efficiency. In this study, an automated method is developed for large-scale production of protein-producing SCs for therapeutic applications. The optimized process, compatible with a robotic liquid handling system (LiHa), reduces production time by half. Additionally, incorporation of an automated tissue dissociator-based emulsification increases batch size 30-fold while preserving SC characteristics. To assess SC quality and protein synthesis, artificial intelligence (AI)-based image analysis is employed, allowing for automated, accurate and high-throughput SC characterization. Large-scale luciferase-expressing SCs from a single homogeneous batch are administered to mice, allowing for real-time monitoring of protein expression and reducing experimental variability. By troubleshooting several central steps in SC synthesis, it is demonstrated that automation and computerized quality control can significantly improve the process of SC synthesis for preclinical and clinical applications.
Collapse
Affiliation(s)
- Noga Sharf-Pauker
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Ido Galil
- Faculty of Computer Science, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Omer Kfir
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Gal Chen
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
- The Interdisciplinary Program for Biotechnology, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Rotem Menachem
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
- Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Jeny Shklover
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| | - Shanny Ackerman
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, 32000, Israel
| |
Collapse
|
6
|
Liu XH, Wang GR, Zhong NN, Wang WY, Liu B, Li Z, Bu LL. Multi-omics in immunotherapy research for HNSCC: present situation and future perspectives. NPJ Precis Oncol 2025; 9:93. [PMID: 40158059 PMCID: PMC11954913 DOI: 10.1038/s41698-025-00886-w] [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: 12/07/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, significantly impacting patient survival and quality of life. The recent emergence of immunotherapy has provided new hope for HNSCC patients, improving survival rates; however, only 15%-20% of patients benefit, and side effects are inevitable. With advancements in omics technologies and the growing prevalence of bioinformatics research, the immune microenvironment of HNSCC has become increasingly well understood, and the molecular mechanisms underlying immunotherapy responses continue to be elucidated. In this review, we summarize commonly used omics techniques and their applications in the research of HNSCC immunotherapy, including predicting and enhancing efficacy, formulating personalized treatment plans, establishing robust preclinical research models, and identifying new immunotherapy targets. Finally, we explore future perspective in terms of sequencing samples, data integration analysis, emerging technologies, clinicopathological features, and interdisciplinary approaches.
Collapse
Affiliation(s)
- Xuan-Hao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Wei-Yu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
- Department of Oral & Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430072, China
| | - Zheng Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, 430071, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430072, China.
- Department of Oral & Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
7
|
Duymaz D, Kebabci AO, Kizilel S. Harnessing the immunomodulatory potential of chitosan and its derivatives for advanced biomedical applications. Int J Biol Macromol 2025; 307:142055. [PMID: 40090654 DOI: 10.1016/j.ijbiomac.2025.142055] [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: 08/22/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
The success of biomaterial applications in medicine, particularly in tissue engineering, relies on achieving a balance between promoting tissue regeneration and controlling the immune response. Due to its natural origin, high biocompatibility, and versatility, chitosan has emerged as a promising biomaterial especially for immunomodulation purposes. Immunomodulation, refers to the deliberate alteration of the immune system's activity to achieve a desired therapeutic effect either by enhancing or suppressing the function of specific immune cells, signaling pathways, or cytokine production. This modulation opens up the unlimited possibilities for the use of biomaterials, especially about the use of natural polymers such as chitosan. Although numerous chitosan-based immunoregulatory strategies have been demonstrated over the past two decades, the lack of in-depth exploration hinders the full potential of strategies that include chitosan and its derivatives in biomedical applications. Thus, in this review, the possible immunomodulatory effects of chitosan, chitosan derivatives and their potential combined with various agents and therapies are investigated in detail. Moreover, this report includes agents for localized immune response control, chitosan-based strategies with complementary immunomodulatory properties to create synergistic effects that will influence the success of cell therapies for enhanced tissue acceptance and regeneration. Finally, the challenges and outlook of chitosan-based therapies as a powerful tool for improving immunomodulatory applications are discussed for paving the way for further studies.
Collapse
Affiliation(s)
- Doğukan Duymaz
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Aybaran O Kebabci
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye.
| |
Collapse
|
8
|
Ren J, Liu K, Hu L, Yang R, Liu Y, Wang S, Chen X, Zhao S, Jing L, Liu T, Hu B, Zhang X, Wang H, Li H. An Efficient Probe-Based Quantitative PCR Assay Targeting Human-Specific DNA in ST6GALNAC3 for the Quantification of Human Cells in Preclinical Animal Models. Mol Biotechnol 2025; 67:1156-1164. [PMID: 38456963 DOI: 10.1007/s12033-024-01115-8] [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: 03/05/2023] [Accepted: 02/04/2024] [Indexed: 03/09/2024]
Abstract
Precise quantification of human cells in preclinical animal models by a sensitive and specific approach is warranted. The probe-based quantitative PCR (qPCR) assay as a sensitive and swift approach is suitable for the quantification of human cells by targeting human-specific DNA sequences. In this study, we developed an efficient qPCR assay targeting human-specific DNA in ST6GALNAC3 (termed ST6GAL-qPCR) for the quantification of human cells in preclinical animal models. ST6GAL-qPCR probe was synthesized with FAM and non-fluorescent quencher-minor groove binder conjugated to the 5' and 3' end of the probe, respectively. Genomic DNA from human, rhesus monkeys, cynomolgus monkeys, New Zealand White rabbits, SD rats, C57BL/6, and BALB/c mice were utilized for analyzing the specificity and sensitivity of the ST6GAL-qPCR assay. The ST6GAL-qPCR assay targeted human-specific DNA was cloned to pUCM-T vector and released by EcoR I/Hind III digestion for generating a calibration curve. Cell mixing experiment was performed to validate the ST6GAL-qPCR assay by analysis of 0.1%, 0.01%, and 0.001% of human leukocytes mixed with murine thymocytes. The ST6GAL-qPCR assay detected human DNA rather than DNA from the tested animal species. The amplification efficiency of the ST6GAL-qPCR assay was 93% and the linearity of calibration curve was R2 = 0.999. The ST6GAL-qPCR assay detected as low as 5 copies of human-specific DNA and is efficient to specially amplify as low as 30-pg human DNA in the presence of 1 μg of DNA from the tested species, respectively. The ST6GAL-qPCR assay was able to quantify as low as 0.01% of human leukocytes within murine thymocytes. This ST6GAL-qPCR assay can be used as an efficient approach for the quantification of human cells in preclinical animal models.
Collapse
Affiliation(s)
- Jinfeng Ren
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ke Liu
- Department of Gastroenterology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Lang Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ruoning Yang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuting Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Siyu Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xinzhu Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Shuli Zhao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Luyao Jing
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Tiantian Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Bin Hu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Xuefeng Zhang
- Jiangsu Tripod Preclinical Research Laboratories Inc, Nanjing, China
| | - Hui Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Hui Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Department of Pathogenic Biology and Immunology, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, China.
| |
Collapse
|
9
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
Patel D, Reese Koç J, Otegbeye F. Creating a GMP cell processing program: A focus on quality and regulation. Best Pract Res Clin Haematol 2025; 38:101614. [PMID: 40274340 DOI: 10.1016/j.beha.2025.101614] [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: 09/29/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Implementing current Good Manufacturing Practice (GMP) regulations and principles even in early phases of cell-based therapy studies is crucial for ensuring safety and reproducible quality of these products. This paper outlines the comprehensive steps necessary to establish a robust GMP-compliant cell processing program in academic programs with emphases on adherence to regulatory and quality standards. While there are different regulatory agencies governing practice across the globe, the prevailing quality principles described here incorporate common requirements and guidelines from agencies such as the United States Food and Drug Administration (FDA) and the European Medicines Agency (EMA). The goal of this review is to provide guidance for developing a quality management program (QMP) that addresses all critical factors impacting each step in the cell therapy product lifecycle: from procurement and receipt of starter material, through manufacturing, testing, storage, distribution, and administration. The QMP should be designed to assure quality outcomes by maintaining qualified and trained staff at all levels as applicable to their job functions; establishing clear policies and procedures; ensuring the qualification of facilities and equipment; using qualified materials for human use; and providing a framework for detection of trends and implementing process improvement.
Collapse
Affiliation(s)
- Darshan Patel
- Therapeutic Products Program, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA.
| | - Jane Reese Koç
- Cellular Therapy Service, Seidman Cancer Center Wesley Immunotherapy Center, 2103 Cornell Road, Cleveland, OH, 44106, USA.
| | - Folashade Otegbeye
- Translational Science and Therapeutics Division, Therapeutic Products Program, Fred Hutchinson Cancer Center, 1100 Fairview Ave. N, Seattle, WA, 98109, USA.
| |
Collapse
|
11
|
Chugh RM, Bhanja P, Zitter R, Gunewardena S, Badkul R, Saha S. Modulation of β-Catenin promotes WNT expression in macrophages and mitigates intestinal injury. Cell Commun Signal 2025; 23:78. [PMID: 39934819 PMCID: PMC11818365 DOI: 10.1186/s12964-025-02065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Macrophages are the major source of WNT ligands. However, the regulation of WNT expression in macrophages has not been studied. In the present study, we have discovered that activation of canonical β-Catenin signaling suppresses WNT expression in macrophages. EVs from these pre-conditioned macrophages promoted intestinal stem cell regeneration and mitigated intestinal injury. METHOD ChIP-seq analysis and validation studies using recombinant DNA construct expressing Luciferase reporter under WNT promoter (e.g. WNT5a and WNT9b) were conducted to demonstrate the involvement of β-Catenin in the transcriptional regulation of WNT expression. The regulatory role of β-Catenin in WNT expression in macrophages was examined by treating these cells with a Tankyrase inhibitor. In addition, the gene expressing β-Catenin was deleted in macrophages using Csf1r.iCre; Ctnnb1fl/fl mice model. Both pharmacological and genetically modulated macrophages were examined for WNT expression and activity by qPCR and TCF/LEF luciferase assay respectively. Additionally, Csf1r.iCre; Ctnnb1fl/fl mice were exposed to irradiation to compare the radiosensitivity with their wildtype littermate. Extracellular vesicles (EVs) were isolated from pre-conditioned WNT-enriched macrophages and infused in irradiated C57BL/6 and Lgr5/eGFP-IRES-Cre-ERT2; R26-ACTB-tdTomato-EGFP mice to determine the regenerative response of intestinal stem cell (ISC) and epithelial repair. Regenerative effects of EVs were also examined in mice model DSS induced colitis. RESULT ChIP-seq analysis and subsequent validation study suggested physical association of β-Catenin with WNT promoters to suppress WNT expression. Macrophage specific deletion of gene expressing β-Catenin or pharmacological inhibition of Tankyrase improves the WNT expression in macrophages several folds compared to control. Transfusion of these preconditioned macrophages or EVs from these cells delivers optimum level of morphogenic WNT to injured epithelium, activates ISC regeneration and mitigated radiation induced intestinal injury. Intestinal epithelium in Csf1r.iCre; Ctnnb1fl/fl mice also showed radioresistance compared to wild type littermate. Moreover, EVs derived from WNT enriched macrophages can mitigate intestinal injury in mice model of DSS induced acute colitis. CONCLUSION The study provides substantial evidence that macrophage-targeted modulation of canonical WNT signaling induces WNT expression in macrophages. Treatment with preconditioned macrophage derived WNT-enriched EVs can be a promising therapeutic approach against intestinal injury.
Collapse
Affiliation(s)
- Rishi Man Chugh
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Payel Bhanja
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ryan Zitter
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sumedha Gunewardena
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rajeev Badkul
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Subhrajit Saha
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
12
|
Wulfse M, Vervoorn MT, Amelink JJGJ, Ballan EM, de Jager SCA, Sluijter JPG, Doevendans PA, Zwetsloot PPM, Van der Kaaij NP. Past Trends and Future Directions of Cardiac Regenerative Medicine - A Systematic Analysis of Clinical Trial Registries. J Cardiovasc Transl Res 2025; 18:209-220. [PMID: 39361114 PMCID: PMC11885401 DOI: 10.1007/s12265-024-10563-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/12/2024] [Indexed: 03/09/2025]
Abstract
Cell therapy, gene therapy, and tissue engineering have been explored as potential strategies to repair or regenerate damaged cardiac tissue. Despite the presence of encouraging preclinical data, clinical trials of regenerative cardiac therapies have yielded mixed results. Our study aimed to investigate the fate of all registered clinical trials within regenerative cardiac medicine, with the purpose of exploring the potential role of publication bias (or trial-completion bias), how published and unpublished research affects the field, and to draw lessons and recommendations for future clinical trials. In this analysis, we show that only a third of all registered trials has yielded results and that a significant number of trials are not completed. Furthermore, we identified significant heterogeneity in study design, study phase, funding, specific therapies used, primary outcome measures and methods of outcome assessment. These observations might hinder the successful translation of cardiac regenerative therapies into clinical practice.
Collapse
Affiliation(s)
- Maaike Wulfse
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Mats T Vervoorn
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Jantijn J G J Amelink
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Elisa M Ballan
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia C A de Jager
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, Regenerative Medicine Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Pieter A Doevendans
- Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter-Paul M Zwetsloot
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Niels P Van der Kaaij
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands.
| |
Collapse
|
13
|
Bradshaw KJ, Leipzig ND. Applications of Regenerative Tissue-Engineered Scaffolds for Treatment of Spinal Cord Injury. Tissue Eng Part A 2025; 31:108-125. [PMID: 39556330 DOI: 10.1089/ten.tea.2024.0194] [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] [Indexed: 11/19/2024] Open
Abstract
Tissue engineering provides a path forward for emerging personalized medicine therapies as well as the ability to bring about cures for diseases or chronic injuries. Traumatic spinal cord injuries (SCIs) are an example of a chronic injury in which no cure or complete functional recovery treatment has been developed. In part, this has been due to the complex and interconnected nature of the central nervous system (CNS), the cellular makeup, its extracellular matrix (ECM), and the injury site pathophysiology. One way to combat the complex nature of an SCI has been to create functional tissue-engineered scaffolds that replace or replenish the aspects of the CNS and tissue/ECM that are damaged following the immediate injury and subsequent immune response. This can be achieved by employing the tissue-engineering triad consisting of cells, biomaterial(s), and environmental factors. Stem cells, with their innate ability to proliferate and differentiate, are a common choice for cellular therapies. Natural or synthetic biomaterials that have tunable characteristics are normally used as the scaffold base. Environmental factors can range from drugs to growth factors (GFs) or proteins, depending on if the idea would be to stimulate exogeneous or endogenous cell populations or just simply retain cells on the scaffold for effective transplantation. For functional regeneration and integration for SCI, the scaffold must promote neuroprotection and neuroplasticity. Tissue-engineering strategies have shown benefits including neuronal differentiation, axonal regeneration, axonal outgrowth, integration into the native spinal cord, and partial functional recovery. Overall, this review focuses on the background that causes SCI to be so difficult to treat, the individual components of the tissue-engineering triad, and how combinatorial scaffolds can be beneficial toward the prospects of future SCI recovery.
Collapse
Affiliation(s)
- Katherine J Bradshaw
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
| | - Nic D Leipzig
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio, USA
| |
Collapse
|
14
|
Sousa AC, Alvites R, Lopes B, Sousa P, Moreira A, Coelho A, Santos JD, Atayde L, Alves N, Maurício AC. Three-Dimensional Printing/Bioprinting and Cellular Therapies for Regenerative Medicine: Current Advances. J Funct Biomater 2025; 16:28. [PMID: 39852584 PMCID: PMC11765675 DOI: 10.3390/jfb16010028] [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: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
The application of three-dimensional (3D) printing/bioprinting technologies and cell therapies has garnered significant attention due to their potential in the field of regenerative medicine. This paper aims to provide a comprehensive overview of 3D printing/bioprinting technology and cell therapies, highlighting their results in diverse medical applications, while also discussing the capabilities and limitations of their combined use. The synergistic combination of 3D printing and cellular therapies has been recognised as a promising and innovative approach, and it is expected that these technologies will progressively assume a crucial role in the treatment of various diseases and conditions in the foreseeable future. This review concludes with a forward-looking perspective on the future impact of these technologies, highlighting their potential to revolutionize regenerative medicine through enhanced tissue repair and organ replacement strategies.
Collapse
Affiliation(s)
- Ana Catarina Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
- Instituto Universitário de Ciências da Saúde (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, Gandra, 4585-116 Paredes, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Alícia Moreira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - André Coelho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - José Domingos Santos
- REQUIMTE-LAQV, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, UP, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal;
| | - Luís Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic Institute of Leiria, Rua de Portugal—Zona Industrial, 2430-028 Marinha Grande, Portugal;
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| |
Collapse
|
15
|
Wu RW, Lin YH, Lu CH, Su CH, Chen YS, Wang FS, Lian WS. Gold nanomaterials capped with bovine serum albumin for cell and extracellular vesicle imaging. NANOTECHNOLOGY 2025; 36:105101. [PMID: 39780321 DOI: 10.1088/1361-6528/ada3da] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025]
Abstract
Bovine serum albumin-capped gold nanoclusters (AuNC@BSA) are ionic, ultra-small, and eco-friendly nanomaterials that exhibit red fluorescence emission. Upon modification, these nanomaterials can serve as imaging probes with multimodal functionality. Owing to their nanoscale properties, AuNC@BSA-based nanomaterials can be readily endocytosed by cells for imaging. With the increasing interest in cell therapy, extracellular vesicles (EVs) have attracted considerable attention from researchers; however, effective methods for imaging EVs remain limited. Although several studies have explored imaging strategies for cells and EVs using compounds, nuclear pharmaceuticals, nanoparticles, or genetic constructs, the use of AuNC@BSA-based nanomaterials for labeling EVs and their parental cells has rarely been discussed, with even less attention paid to their multimodal potential. To address this gap, we utilized three types of AuNC@BSA-based derivatives: AuNC@BSA, AuNC@BSA-Gd, and AuNC@BSA-Gd-I. Our findings demonstrate that these derivatives can effectively label both cells and EVs using a simple direct labeling approach, which is particularly notable for EVs, as they typically require more complex labeling procedures. Furthermore, the multimodal potential of labeled cells and EVs was evaluated, revealing their capabilities for multimodal imaging. In summary, this study presents an effective strategy for labeling EVs and their parental cells using multimodal nanomaterials. These findings will contribute to accelerating the development of drug delivery systems, cell- and EV-based therapies, and advanced imaging strategies.
Collapse
Affiliation(s)
- Re-Wen Wu
- Department of Orthopedic Surgery, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Yu-Han Lin
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Cheng-Hsiu Lu
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Chia-Hao Su
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Center for General Education, Chang Gung University, Taoyuan 333, Taiwan
- Institute for Radiological Research, Chang Gung University, Taoyuan 333, Taiwan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Shan Chen
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Feng-Sheng Wang
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Wei-Shiung Lian
- Center for Mitochondrial Research and Medicine, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Department of Medical Research, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Core Laboratory for Phenomics and Diagnostic, College of Medicine Chang Gung University, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
16
|
Yin GN, Ryu JK. Role of pericytes in regulating penile angiogenesis and nerve regeneration. Asian J Androl 2025; 27:13-19. [PMID: 39162179 PMCID: PMC11784945 DOI: 10.4103/aja202455] [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: 02/18/2024] [Accepted: 05/19/2024] [Indexed: 08/21/2024] Open
Abstract
ABSTRACT Pericytes are multifunctional mural cells that surround the abluminal wall of endothelial cells and are associated with vascular development, vascular permeability, and angiogenesis. Additionally, pericytes demonstrate stem cell-like properties and contribute to neuroinflammatory processes. Pericytes have been extensively studied in the central nervous system. However, specific mechanisms underlying its involvement in various physiological and pathological conditions, especially in erectile dysfunction (ED), remain poorly understood. Advancements in in vitro and in vitro techniques, such as single-cell RNA sequencing, are expanding our understanding of pericytes. Recent studies have shown that pericyte dysfunction is considered an important factor in the pathogenesis of vascular and neurological ED. Therefore, this study aims to analyze the specific role of pericytes in ED, focusing on diabetic and neurogenic ED. This article provides a comprehensive review of research findings on PubMed from 2000 to 2023, concerning pericyte dysfunction in the process of ED, offering valuable insights, and suggesting directions for further research.
Collapse
Affiliation(s)
- Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon 22332, Korea
- Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| |
Collapse
|
17
|
Var SR, Strell P, Shetty A, Roman A, Clark IH, Crane AT, Dunbar GL, Fink K, Grande AW, Parr AM, Rossignol J, Sanberg PR, Zhao LR, Zholudeva LV, Low WC. Research Guideline Recommendations for Research on Stem Cells, Human Embryos, and Gene Editing. Cell Transplant 2025; 34:9636897241312793. [PMID: 40007211 PMCID: PMC11863228 DOI: 10.1177/09636897241312793] [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: 09/03/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 02/27/2025] Open
Abstract
Recent advances in biomedical technologies have extended the boundaries of previously established regulatory guidelines pertaining to stem cell research. These guidelines constrained the study of human pluripotent stem cells (hPSCs) and their derivatives from use under various conditions, including the introduction of hPSCs into the brains of host animals because of concerns of humanizing the brains of animal species. Other guidelines constrained the use of hPSCs in creating human-animal chimeras because of the potential contribution of human stem cells not only to the brain but also to the germline. Some regulatory guidelines forbid the growing of human embryos ex vivo beyond the stage of primitive streak development because of concerns regarding the creation of human forms of life ex vivo. At the subcellular level, there are guidelines regulating the transfer of mitochondria within human embryos. At the molecular level, there are guidelines regulating genome editing to prevent permanent genetic alterations in germline cells. These and other issues related to stem cells have been reviewed, and new research guidelines established by the International Society for Stem Cell Research (ISSCR) for its membership. Because many of the recommended changes by the ISSCR impact research being conducted by members of the American Society for Neural Therapy and Repair (ASNTR), the ASNTR established a task force to review relevant recommendations by the ISSCR to determine which new guidelines to adopt for research conducted by the ASNTR society membership. The final ASNTR recommendations are presented in this document.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Phoebe Strell
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Comparative and Molecular Biosciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
| | - Anala Shetty
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
| | - Alex Roman
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Isaac H. Clark
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Andrew T. Crane
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Gary L. Dunbar
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, USA
| | - Kyle Fink
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Ann M. Parr
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Julien Rossignol
- College of Medicine, Central Michigan University, Mount Pleasant, MI, USA
| | - Paul R. Sanberg
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
| | - Li-Ru Zhao
- Department of Neurosurgery, The State University of New York Upstate Medical University, Syracuse, NY, USA
| | | | - Walter C. Low
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
- Department of Comparative and Molecular Biosciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, USA
- Department of Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, MN, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
18
|
Fernandes Q, Folorunsho OG. Unveiling the nexus: The tumor microenvironment as a strategic frontier in viral cancers. Cytokine 2025; 185:156827. [PMID: 39647395 DOI: 10.1016/j.cyto.2024.156827] [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: 09/28/2024] [Revised: 11/23/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Viral infections are a significant factor in the etiology of various cancers, with the tumor microenvironment (TME) playing a crucial role in disease progression. This review delves into the complex interactions between viruses and the TME, highlighting how these interactions shape the course of viral cancers. We explore the distinct roles of immune cells, including T-cells, B-cells, macrophages, and dendritic cells, within the TME and their influence on cancer progression. The review also examines how viral oncoproteins manipulate the TME to promote immune evasion and tumor survival. Unraveling these mechanisms highlights the emerging paradigm of targeting the TME as a novel approach to cancer treatment. Our analysis provides insights into the dynamic interplay between viruses and the TME, offering a roadmap for innovative treatments that leverage the unique characteristics of viral cancers.
Collapse
Affiliation(s)
- Queenie Fernandes
- College of Medicine, QU Health, Qatar University, Doha 2713, Qatar; Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, PO. Box 3050, Doha, Qatar.
| | - Oginni Gbenga Folorunsho
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska cesta 5000, Nova Gorica, Slovenia
| |
Collapse
|
19
|
Murugaiyan K, Murali VP, Tamura H, Furuike T, Rangasamy J. Overview of chitin dissolution, hydrogel formation and its biomedical applications. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-32. [PMID: 39704399 DOI: 10.1080/09205063.2024.2442181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Chitin hydrogel and hydrogel-based products are some of the frequently reported biomaterials for biomedical applications. Yet there is a void in understanding chitin's dissolution mechanism and its most suitable solvent system(s). Chitin is a natural polysaccharide polymer which can be dissolved in solvents such as calcium chloride- methanol, sodium hydroxide/urea (NaOH/urea), lithium chloride diacetamide (LiCl/DMAc), ionic liquids and deep eutectic solvents. Among the alkali/urea dissolution systems such as NaOH/urea, KOH/urea, LiOH/urea for dissolution of chitin we will be focussing on NaOH-based system here for ease of comparison with the other systems. Chitin has been used for decades in the biomedical field; however, new solvent systems are still being explored even to this day to identify the most suitable chemical(s) for dissolving it. Chitin, due to its biocompatibility, allows us to use it for multifaceted purposes. Hence, it is important to consolidate the available studies for better understanding about the most sought-after biomaterial. This overview deeply delves into the mechanism of action of the existing solvent systems and highlights its merits and demerits. It discusses the rheological properties of the chitin gel from different solvent systems and puts forth the current biomedical applications of chitin gel in areas such as tissue engineering, drug delivery, biosensing, hemostasis and wound healing. It also outlines recent advances and highlights the potential gaps which need to be addressed in future studies.
Collapse
Affiliation(s)
- Kavipriya Murugaiyan
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| | | | - Hiroshi Tamura
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
- Organization for Research and Development of Innovative Science and Technology (ORDIST), Kansai University, Suita, Osaka, Japan
| | - Tetsuya Furuike
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
- Organization for Research and Development of Innovative Science and Technology (ORDIST), Kansai University, Suita, Osaka, Japan
| | - Jayakumar Rangasamy
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, India
| |
Collapse
|
20
|
Medico F, Kim S, Surwase SS, Liu H, Kim YC. A core-shell microneedle system for stable fibroblast delivery in cell-based therapies. Drug Deliv Transl Res 2024:10.1007/s13346-024-01759-8. [PMID: 39699751 DOI: 10.1007/s13346-024-01759-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 12/20/2024]
Abstract
Human cells, such as fibroblasts and particularly human mesenchymal stem cells (hMSCs), represent a promising and effective therapeutic tool for a range of cell-based therapies used to treat various diseases. The effective delivery of therapeutic cells remains a challenge due to limitations in targeting, invasiveness, and cell viability. To address these challenges, we developed a microneedle (MN) system for minimally invasive cell delivery with high cellular stability. The MN system comprises a core of gelatin methacryloyl (GelMA) hydrogel embedded with fibroblasts, encased in a polylactic-co-glycolic acid (PLGA) shell that enhances structural integrity for efficient skin penetration. The fabrication process involves UV-crosslinking of the GelMA hydrogel with cells, optimizing both cell encapsulation and structural strength. This MN system achieves over 80% cell viability after seven days in vitro, with the conventional GelMA formulation providing superior stability and cellular outcomes. This platform's ability to ensure sustained cell viability presents promising implications for future applications in regenerative medicine, wound healing, and localized treatments for skin conditions. This MN system opens new avenues for cell-based therapies, offering a versatile and scalable solution for therapeutic cell delivery.
Collapse
Affiliation(s)
- Federica Medico
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seungcheol Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sachin S Surwase
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Haoyan Liu
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.
| |
Collapse
|
21
|
Kutle I, Polten R, Stalp JL, Hachenberg J, Todzey F, Hass R, Zimmermann K, von der Ohe J, von Kaisenberg C, Neubert L, Kamp JC, Schaudien D, Seyda AK, Hillemanns P, Klapdor R, Morgan MA, Schambach A. Anti-Mesothelin CAR-NK cells as a novel targeted therapy against cervical cancer. Front Immunol 2024; 15:1485461. [PMID: 39781381 PMCID: PMC11707549 DOI: 10.3389/fimmu.2024.1485461] [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: 08/23/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
Resistance to the currently available treatment paradigms is one of the main factors that contributes to poor outcomes in patients with advanced cervical cancer. Novel targeted therapy approaches might enhance the patient's treatment outcome and are urgently needed for this malignancy. While chimeric-antigen receptor (CAR)-based adoptive immunotherapy displays a promising treatment strategy for liquid cancers, their use against cervical cancer is largely unexplored. This study used alpharetroviral SIN vectors to equip natural killer (NK) cells with a third-generation CAR (including CD28 and 4-1BB co-stimulatory domains) targeting Mesothelin, which was identified to be highly expressed on primary human cervical cancer tissues and cervical cancer cell lines in this and other studies. Anti-Mesothelin CAR-NK cells demonstrated high cytotoxicity against cervical cancer cells in 2D and 3D culture models, which corresponded to increased degranulation of CAR-NK-92 cells upon exposure to Mesothelin+ target cells. Mesothelin- cervical cancer cells were generated by CRISPR-Cas9-mediated knockout and used to show target antigen specificity of anti-Mesothelin CAR-NK-92 cells and primary NK cells derived from different healthy donors in co-culture experiments. Combination of anti-Mesothelin CAR-NK-92 cells with chemotherapy revealed increased elimination of cancer cells as compared to monotherapy settings. Our findings indicate the promise of anti-Mesothelin CAR-NK cells as a potential treatment option against cervical cancer, as well as other Mesothelin+ malignancies.
Collapse
Affiliation(s)
- Ivana Kutle
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Robert Polten
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Jan Lennart Stalp
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Jens Hachenberg
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Felix Todzey
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Ralf Hass
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Katharina Zimmermann
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Juliane von der Ohe
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | | | - Lavinia Neubert
- Institute of Pathology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Jan C. Kamp
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany
- Department of Respiratory Medicine and Infectious Diseases, Hannover Medical School, Hannover, Germany
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, ITEM, Hannover, Germany
| | - Ann-Kathrin Seyda
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Peter Hillemanns
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Rüdiger Klapdor
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
- Department of Gynecology and Obstetrics, Albertinen Hospital Hamburg, Hamburg, Germany
| | | | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Division of Hematology/Oncology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
22
|
Asadi Sarabi P, Shabanpouremam M, Eghtedari AR, Barat M, Moshiri B, Zarrabi A, Vosough M. AI-Based solutions for current challenges in regenerative medicine. Eur J Pharmacol 2024; 984:177067. [PMID: 39454850 DOI: 10.1016/j.ejphar.2024.177067] [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: 09/08/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
The emergence of Artificial Intelligence (AI) and its usage in regenerative medicine represents a significant opportunity that holds the promise of tackling critical challenges and improving therapeutic outcomes. This article examines the ways in which AI, including machine learning and data fusion techniques, can contribute to regenerative medicine, particularly in gene therapy, stem cell therapy, and tissue engineering. In gene therapy, AI tools can boost the accuracy and safety of treatments by analyzing extensive genomic datasets to target and modify genetic material in a precise manner. In cell therapy, AI improves the characterization and optimization of cell products like mesenchymal stem cells (MSCs) by predicting their function and potency. Additionally, AI enhances advanced microscopy techniques, enabling accurate, non-invasive and quantitative analyses of live cell cultures. AI enhances tissue engineering by optimizing biomaterial and scaffold designs, predicting interactions with tissues, and streamlining development. This leads to faster and more cost-effective innovations by decreasing trial and error. The convergence of AI and regenerative medicine holds great transformative potential, promising effective treatments and innovative therapeutic strategies. This review highlights the importance of interdisciplinary collaboration and the continued integration of AI-based technologies, such as data fusion methods, to overcome current challenges and advance regenerative medicine.
Collapse
Affiliation(s)
- Pedram Asadi Sarabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahshid Shabanpouremam
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Faculty of Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Amir Reza Eghtedari
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Mahsa Barat
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Behzad Moshiri
- School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan, 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
23
|
Arjmand B, Alavi-Moghadam S, Khorsand G, Sarvari M, Arjmand R, Rezaei-Tavirani M, Rajaeinejad M, Mosaed R. Cell-Based Vaccines: Frontiers in Medical Technology for Cancer Treatment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2024; 10:480-499. [DOI: 10.1007/s40883-024-00338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 01/13/2024] [Accepted: 02/17/2024] [Indexed: 01/03/2025]
|
24
|
Kahts M, Mellet J, Durandt C, Moodley K, Summers B, Ebenhan T, Zeevaart JR, Aras O, Pepper MS. A proof-of-concept study to investigate the radiolabelling of human mesenchymal and hematopoietic stem cells with [ 89Zr]Zr-Df-Bz-NCS. EJNMMI Radiopharm Chem 2024; 9:82. [PMID: 39611856 PMCID: PMC11607195 DOI: 10.1186/s41181-024-00311-w] [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: 10/12/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND The transplantation of hematopoietic stem and progenitor cells (HSPCs) or mesenchymal stromal/stem cells (MSCs) for the treatment of a wide variety of diseases has been studied extensively. A challenge with cell-based therapies is that migration to and retention at the target site is often difficult to monitor and quantify. Zirconium-89 (89Zr) is a positron-emitting radionuclide with a half-life of 3.3 days, which allows long-term cell tracking. Para-isothiocyanatobenzyl-desferrioxamine B (Df-Bz-NCS) is the chelating agent of choice for 89Zr-cell surface labelling. We utilised a shortened labelling method, thereby avoiding a 30-60-min incubation step for [89Zr]Zr-Df-Bz-NCS chelation, to radiolabel HSPCs and MSCs with zirconium-89. RESULTS Three 89Zr-MSC labelling attempts were performed. High labelling efficiencies (81.30 and 87.30%) and relatively good labelling yields (59.59 and 67.00%) were achieved with the use of a relatively larger number of MSCs (4.425 and 3.855 million, respectively). There was no significant decrease in MSC viability after 89Zr-labeling (p = 0.31). This labelling method was also translatable to prepare 89Zr-HSPC; preliminary data from one preparation indicated high 89Zr-HSPC labelling efficiency (88.20%) and labelling yield (71.06%), as well as good HSPC viability after labelling (68.65%). CONCLUSIONS Successful 89Zr-MSC and 89Zr-HSPC labelling was achieved, which underlines the prospects for in vivo cell tracking studies with positron emission tomography. In vitro investigations with larger sample sizes and preclinical studies are recommended.
Collapse
Affiliation(s)
- Maryke Kahts
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa.
| | - Juanita Mellet
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Chrisna Durandt
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Kinosha Moodley
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Beverley Summers
- Pharmaceutical Sciences Department, School of Pharmacy, Sefako Makgatho Health Sciences University (SMU), Ga-Rankuwa, 0208, South Africa
| | - Thomas Ebenhan
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Hartebeespoort, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- DST/NWU, Preclinical Drug Development Platform, North-West University, Potchefstroom, South Africa
| | - Omer Aras
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Radiology Department, AMRIC Health, New York, NY, USA
| | - Michael S Pepper
- Department of Medical Immunology, Institute for Cellular and Molecular Medicine, South African Medical Research Council (SAMRC) Extramural Unit for Stem Cell Research and Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
25
|
Gurdal M, Ercan G, Barut Selver O, Aberdam D, Zeugolis DI. Development of Biomimetic Substrates for Limbal Epithelial Stem Cells Using Collagen-Based Films, Hyaluronic Acid, Immortalized Cells, and Macromolecular Crowding. Life (Basel) 2024; 14:1552. [PMID: 39768260 PMCID: PMC11678493 DOI: 10.3390/life14121552] [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: 10/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 01/05/2025] Open
Abstract
Despite the promising potential of cell-based therapies developed using tissue engineering techniques to treat a wide range of diseases, including limbal stem cell deficiency (LSCD), which leads to corneal blindness, their commercialization remains constrained. This is primarily attributable to the limited cell sources, the use of non-standardizable, unscalable, and unsustainable techniques, and the extended manufacturing processes required to produce transplantable tissue-like surrogates. Herein, we present the first demonstration of the potential of a novel approach combining collagen films (CF), hyaluronic acid (HA), human telomerase-immortalized limbal epithelial stem cells (T-LESCs), and macromolecular crowding (MMC) to develop innovative biomimetic substrates for limbal epithelial stem cells (LESCs). The initial step involved the fabrication and characterization of CF and CF enriched with HA (CF-HA). Subsequently, T-LESCs were seeded on CF, CF-HA, and tissue culture plastic (TCP). Thereafter, the effect of these matrices on basic cellular function and tissue-specific extracellular matrix (ECM) deposition with or without MMC was evaluated. The viability and metabolic activity of cells cultured on CF, CF-HA, and TCP were found to be similar, while CF-HA induced the highest (p < 0.05) cell proliferation. It is notable that CF and HA induced cell growth, whereas MMC increased (p < 0.05) the deposition of collagen IV, fibronectin, and laminin in the T-LESC culture. The data highlight the potential of, in particular, immortalized cells and MMC for the development of biomimetic cell culture substrates, which could be utilized in ocular surface reconstruction following further in vitro, in vivo, and clinical validation of the approach.
Collapse
Affiliation(s)
- Mehmet Gurdal
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, 35100 Izmir, Türkiye;
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland;
| | - Gulinnaz Ercan
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, 35100 Izmir, Türkiye;
- Department of Stem Cell, Institute of Health Sciences, Ege University, 35100 Izmir, Türkiye;
| | - Ozlem Barut Selver
- Department of Stem Cell, Institute of Health Sciences, Ege University, 35100 Izmir, Türkiye;
- Department of Ophthalmology, Faculty of Medicine, Ege University, 35100 Izmir, Türkiye
| | - Daniel Aberdam
- INSERM U1138, Centre des Cordeliers, Université de Paris, 75006 Paris, France;
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, H91 TK33 Galway, Ireland;
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| |
Collapse
|
26
|
Son KH, Kim DH, Park S, Kim HJ, Park M, Kim SJ, Lee SJ, Ahn K, Lee JW. Spherical Shell Bioprinting to Produce Uniform Spheroids with Controlled Sizes. J Funct Biomater 2024; 15:350. [PMID: 39590553 PMCID: PMC11595458 DOI: 10.3390/jfb15110350] [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: 11/01/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Conventional cell spheroid production methods are largely manual, leading to variations in size and shape that compromise consistency and reliability for use in cell-based therapeutic applications. To enhance spheroid production, a spherical shell bioprinting system was implemented, enabling the high-throughput generation of uniform cell spheroids with precisely controlled sizes. The system encapsulates cells within thin alginate hydrogel shells formed through bioprinting and ion crosslinking reactions. Alginate-calcium ion crosslinking created alginate shells that contained gelatin-based bioinks with embedded cells, facilitating spontaneous cell aggregation within the shells and eliminating the need for plastic wells. By adjusting cell concentrations in the alginate-gelatin bioink, we achieved precise control over spheroid size, maintaining a sphericity above 0.94 and size deviations within ±10 µm. This method has been successfully applied to various cell types including cancer cells, fibroblasts, chondrocytes, and epithelial cells, demonstrating its versatility. This scalable approach enhances the reliability of cell therapy and drug screening, offering a robust platform for future biomedical applications.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, College of Medicine, Gachon University, 21, Namdong-daero 774 Beon-gil, Namdong-gu, Incheon 21565, Republic of Korea;
| | - Dong-Ha Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seunghye Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
| | - Hyun Jae Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| |
Collapse
|
27
|
Kim S, Sharma C, Hong J, Kim JH, Nam Y, Kim MS, Lee TY, Kim KS, Suk K, Lee HW, Kim SR. Post-symptomatic administration of hMSCs exerts therapeutic effects in SCA2 mice. Stem Cell Res Ther 2024; 15:411. [PMID: 39521966 PMCID: PMC11550562 DOI: 10.1186/s13287-024-04020-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Defects in the ataxin-2 (ATXN-2) protein and CAG trinucleotide repeat expansion in its coding gene, Atxn-2, cause the neurodegenerative disorder spinocerebellar ataxia type 2 (SCA2). While clinical studies suggest potential benefits of human-derived mesenchymal stem cells (hMSCs) for treating various ataxias, the exact mechanisms underlying their therapeutic effects and interaction with host tissue to stimulate neurotrophin expression remain unclear specifically in the context of SCA2. METHODS Human bone marrow-derived MSCs (hMSCs) were injected into the cisterna magna of 26-week-old wild-type and SCA2 mice. Mice were assessed for impaired motor coordination using the accelerating rotarod, open field test, and composite phenotype scoring. At 50 weeks, the cerebellum vermis was harvested for protein assessment and immunohistochemical analysis. RESULTS Significant loss of NeuN and calbindin was observed in 25-week-old SCA2 mice. However, after receiving multiple injections of hMSCs starting at 26 weeks of age, these mice exhibited a significant improvement in abnormal motor performance and a protective effect on Purkinje cells. This beneficial effect persisted until the mice reached 50 weeks of age, at which point they were sacrificed to study further mechanistic events triggered by the administration of hMSCs. Calbindin-positive cells in the Purkinje cell layer expressed bone-derived neurotrophic factor after hMSC administration, contributing to the protection of cerebellar neurons from cell death. CONCLUSION In conclusion, repeated administration of hMSCs shows promise in alleviating SCA2 symptoms by preserving Purkinje cells, improving neurotrophic support, and reducing inflammation, ultimately leading to the preservation of locomotor function in SCA2 mice.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Chanchal Sharma
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea
- Byrd Alzheimer's Centre and Research Institute, University of South Florida, Tampa, FL, 33620, USA
| | - Jungwan Hong
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Jong-Heon Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
| | - Min Sung Kim
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Tae Yong Lee
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Kyung-Suk Kim
- Bioengineering Institute, Corestemchemon Inc, Seoul, 13486, Korea
| | - Kyoungho Suk
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
- Department of Pharmacology and Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Korea
| | - Ho-Won Lee
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea
- Department of Neurology, Kyungpook National University Chilgok Hospital, Daegu, 41404, Korea
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, 41566, Korea.
- Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41944, Korea.
| |
Collapse
|
28
|
Celik B, Rintz E, Sansanwal N, Khan S, Bigger B, Tomatsu S. Lentiviral Vector-Mediated Ex Vivo Hematopoietic Stem Cell Gene Therapy for Mucopolysaccharidosis IVA Murine Model. Hum Gene Ther 2024; 35:917-937. [PMID: 39446675 DOI: 10.1089/hum.2024.094] [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: 10/26/2024] Open
Abstract
Mucopolysaccharidosis IVA (MPS IVA) is an autosomal recessive disease caused by a mutation in the N-acetylgalactosamine-6-sulfate-sulfatase (GALNS) gene resulting in progressive systemic skeletal dysplasia. There is currently no effective treatment available for this skeletal condition. Thus, the development of a new therapy stands as an unmet challenge in reversing or alleviating the progression of the disease. Our research, which could be a game-changer, hypothesizes that ex vivo lentiviral (LV) gene therapy (GT) could produce the supraphysiological level of active GALNS enzyme by hematopoietic stem cells (HSCs) transduced with LVs carrying the native GALNS gene under two different promoters (CBh and COL2A1), impacting bone and cartilage abnormalities in MPS IVA. We conditioned newborn knock-out (Galns-/-) MPS IVA mice with busulfan and intravenously transplanted LV-modified HSCs isolated from the bone marrow of Galns-/- donor mice. Transplanted mice were autopsied at 16 weeks, and tissues were collected to assess the therapeutic efficacy of modified HSCs in MPS IVA mice. Although HSC-LV-CBh-hGALNS provided a higher GALNS enzyme activity in plasma, HSC-LV-COL2A1-hGALNS stably corrected heart and bone abnormalities better under a low level of GALNS enzyme. Our findings suggest that ex vivo LV-GT may potentially treat MPS IVA.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
- Skeletal Dysplasia Research Lab, Nemours Children's Health, Wilmington, Delaware, USA
| | - Estera Rintz
- Skeletal Dysplasia Research Lab, Nemours Children's Health, Wilmington, Delaware, USA
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Nidhi Sansanwal
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
- Skeletal Dysplasia Research Lab, Nemours Children's Health, Wilmington, Delaware, USA
| | - Shaukat Khan
- Skeletal Dysplasia Research Lab, Nemours Children's Health, Wilmington, Delaware, USA
| | - Brian Bigger
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Shunji Tomatsu
- Skeletal Dysplasia Research Lab, Nemours Children's Health, Wilmington, Delaware, USA
| |
Collapse
|
29
|
Hegazi A, Rager LE, Watkins DE, Su KH. Advancing Immunotherapy in Pancreatic Cancer. Int J Mol Sci 2024; 25:11560. [PMID: 39519112 PMCID: PMC11546161 DOI: 10.3390/ijms252111560] [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: 10/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic cancer remains one of the deadliest malignancies, with a consistently low five-year survival rate for the past several decades. This is in stark contrast to other cancers, which have seen significant improvement in survival and prognosis due to recent developments in therapeutic modalities. These modest improvements in pancreatic cancer outcomes have primarily resulted from minor advances in cytotoxic chemotherapeutics, with limited progress in other treatment approaches. A major focus of current therapeutic research is the further development of immunomodulatory therapies characterized by antibody-based approaches, cellular therapies, and vaccines. Although initial results utilizing immunotherapy in pancreatic cancer have been mixed, recent clinical trials have demonstrated significant improvements in patient outcomes. In this review, we detail these three approaches to immunomodulation, highlighting their common targets and distinct shortcomings, and we provide a narrative summary of completed and ongoing clinical trials that utilize these approaches to immunomodulation. Within this context, we aim to inform future research efforts by identifying promising areas that warrant further exploration.
Collapse
Affiliation(s)
| | | | | | - Kuo-Hui Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA; (A.H.); (L.E.R.); (D.E.W.)
| |
Collapse
|
30
|
Chen H, Bao Y, Li X, Chen F, Sugimura R, Zeng X, Xia J. Cell Surface Engineering by Phase-Separated Coacervates for Antibody Display and Targeted Cancer Cell Therapy. Angew Chem Int Ed Engl 2024; 63:e202410566. [PMID: 39103291 DOI: 10.1002/anie.202410566] [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: 06/04/2024] [Revised: 07/16/2024] [Accepted: 08/04/2024] [Indexed: 08/07/2024]
Abstract
Cell therapies such as CAR-T have demonstrated significant clinical successes, driving the investigation of immune cell surface engineering using natural and synthetic materials to enhance their therapeutic performance. However, many of these materials do not fully replicate the dynamic nature of the extracellular matrix (ECM). This study presents a cell surface engineering strategy that utilizes phase-separated peptide coacervates to decorate the surface of immune cells. We meticulously designed a tripeptide, Fmoc-Lys-Gly-Dopa-OH (KGdelta; Fmoc=fluorenylmethyloxycarbonyl; delta=Dopa, dihydroxyphenylalanine), that forms coacervates in aqueous solution via phase separation. These coacervates, mirroring the phase separation properties of ECM proteins, coat the natural killer (NK) cell surface with the assistance of Fe3+ ions and create an outer layer capable of encapsulating monoclonal antibodies (mAb), such as Trastuzumab. The antibody-embedded coacervate layer equips the NK cells with the ability to recognize cancer cells and eliminate them through enhanced antibody-dependent cellular cytotoxicity (ADCC). This work thus presents a unique strategy of cell surface functionalization and demonstrates its use in displaying cancer-targeting mAb for cancer therapies, highlighting its potential application in the field of cancer therapy.
Collapse
Affiliation(s)
- Hongfei Chen
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, 99999, Hong Kong SAR, China
| | - Yishu Bao
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, 99999, Hong Kong SAR, China
| | - Xiaojing Li
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, 99999, Hong Kong SAR, China
| | - Fangke Chen
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, 99999, Hong Kong SAR, China
| | - Ryohichi Sugimura
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, 99999, Hong Kong SAR, China
| | - Xiangze Zeng
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, 99999, Hong Kong SAR, China
| | - Jiang Xia
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, 99999, Hong Kong SAR, China
| |
Collapse
|
31
|
Lu Y, Shi R, He W, An Q, Zhao J, Gao X, Zhang B, Zhang L, Xu K, Ma D. Cell therapy in Sjögren's syndrome: opportunities and challenges. Expert Rev Mol Med 2024; 26:e28. [PMID: 39438246 PMCID: PMC11505611 DOI: 10.1017/erm.2024.21] [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: 08/22/2023] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 10/25/2024]
Abstract
Sjögren's syndrome (SS) is a chronic autoimmune disease caused by immune system disorders. The main clinical manifestations of SS are dry mouth and eyes caused by the destruction of exocrine glands, such as the salivary and lacrimal glands, and systemic manifestations, such as interstitial pneumonia, interstitial nephritis and vasculitis. The pathogenesis of this condition is complex. However, this has not been fully elucidated. Treatment mainly consists of glucocorticoids, disease-modifying antirheumatic drugs and biological agents, which can only control inflammation but not repair the tissue. Therefore, identifying methods to regulate immune disorders and repair damaged tissues is imperative. Cell therapy involves the transplantation of autologous or allogeneic normal or bioengineered cells into the body of a patient to replace damaged cells or achieve a stronger immunomodulatory capacity to cure diseases, mainly including stem cell therapy and immune cell therapy. Cell therapy can reduce inflammation, relieve symptoms and promote tissue repair and regeneration of exocrine glands such as the salivary glands. It has broad application prospects and may become a new treatment strategy for patients with SS. However, there are various challenges in cell preparation, culture, storage and transportation. This article reviews the research status and prospects of cell therapies for SS.
Collapse
Affiliation(s)
- Yangyang Lu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Rongjing Shi
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Wenqin He
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Qi An
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Jingwen Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Xinnan Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Baiyan Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Ke Xu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
| | - Dan Ma
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Province Clinical Research Center for Dermatologic and Immunologic Diseases (Rheumatic diseases), Taiyuan, China
- Shanxi Province Clinical Theranostics Technology Innovation Center for Immunologic and Rheumatic Diseases, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| |
Collapse
|
32
|
Du X, Luo X, Liu L, Cao Y, Zhang Y, Zhang Y. Mapping the cell therapy landscape: insights into clinical trials and regulatory advances in China. J Hematol Oncol 2024; 17:96. [PMID: 39402668 PMCID: PMC11476068 DOI: 10.1186/s13045-024-01616-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
In recent years, cell therapy research and commercialization have significantly accelerated, especially after the US FDA approved CAR-T therapy. While cell therapy now leads immuno-oncology in clinical trials, challenges such as redundant R&D, target clustering, and unmet clinical need remain. Since 2017, China has established a dual-track regulatory framework, facilitating rapid growth in its cell therapy pipeline, making it the second largest in the world. Despite this progress, China faces similar global challenges. Our study covers 2,794 registered cell therapy clinical trials in China, including 2,045 for immune cell, 683 for stem cell, and 66 for other somatic cell. It compares cell therapy products approved in China, the US, EU, and Japan, analyzes the evolving clinical trials landscape, and highlights the characteristics of investigator-initiated trials (IITs) and industry-sponsored trials (ISTs) in China. Our findings indicate that despite the high disease burden and unmet clinical needs for solid tumors in China, over 38% of trials between 2021 and 2023 focused on hematologic malignancies with established targets like CD19 and BCMA. Over 90% of trials are IITs, which show notable clinical differences from ISTs. We recommend that Chinese regulators establish specific guidelines to promote clinical-value-driven research. Stricter regulatory standards should also be implemented to minimize redundant R&D. Additionally, a value-based reimbursement system for within-class targeted cell therapy products may further reduce duplicated R&D efforts. Given the prevalence of IITs, specifying requirements for IITs could create a new pathway to accelerate product development and better address unmet clinical needs in China.
Collapse
Affiliation(s)
- Xin Du
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Xingxian Luo
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Lanqiu Liu
- School of Medical Humanities, Capital Medical University, Beijing, China
| | - Yanlin Cao
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yajuan Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Key Laboratory of Innovative Drug Research and Evaluation, National Medical Products Administration, Beijing, China.
| | - Yi Zhang
- Vanke School of Public Health, Tsinghua University, Beijing, China.
| |
Collapse
|
33
|
Looi CK, Loo EM, Lim HC, Chew YL, Chin KY, Cheah SC, Goh BH, Mai CW. Revolutionizing the treatment for nasopharyngeal cancer: the impact, challenges and strategies of stem cell and genetically engineered cell therapies. Front Immunol 2024; 15:1484535. [PMID: 39450176 PMCID: PMC11499120 DOI: 10.3389/fimmu.2024.1484535] [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: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a distinct malignancy of the nasopharynx and is consistently associated with the Epstein-Barr virus (EBV) infection. Its unique anatomical location and complex aetiology often result in advanced-stage disease at first diagnosis. While radiotherapy (RT) and chemotherapy have been the mainstays of treatment, they often fail to prevent tumour recurrence and metastasis, leading to high rates of treatment failure and mortality. Recent advancement in cell-based therapies, such as chimeric antigen receptor (CAR)-T cell therapy, have shown great promise in hematological malignancies and are now being investigated for NPC. However, challenges such as targeting specific tumour antigens, limited T cell persistence and proliferation, and managing treatment-related toxicities must be addressed. Extensive research is needed to enhance the effectiveness and safety of these therapies, paving the way for their integration into standard clinical practice for better management of NPC and a better quality of life for human health.
Collapse
Affiliation(s)
- Chin-King Looi
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Ee-Mun Loo
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
- Advanced Genomics Laboratory, AGTC Genomics, Kuala Lumpur, Malaysia
| | - Heng-Chee Lim
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Yik-Ling Chew
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Port Dickson, Negeri Sembilan, Malaysia
| | - Bey Hing Goh
- Sunway Biofunctional Molecules Discovery Centre, School of Medical and Life Sciences, Sunway University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- Biofunctional Molecule Exploratory Research Group, School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor Darul Ehsan, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Zhejiang, China
| | - Chun-Wai Mai
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
34
|
Chakra MA, Bailly H, Klampke F, Boaz J, Jida M, Yassine AA, McElree IM, Moussa M. An update on the use of stem cell therapy for erectile dysfunction. Asian J Urol 2024; 11:530-544. [PMID: 39534008 PMCID: PMC11551375 DOI: 10.1016/j.ajur.2023.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/24/2023] [Indexed: 11/16/2024] Open
Abstract
Objective This systematic review aimed to analyze animal and human trial data to better understand the efficacy of stem cell therapy (SCT) for erectile dysfunction (ED) and the obstacles that may hinder its application in this field. Methods We searched electronic databases, including PubMed and Scopus, for published studies with the Medical Subject Heading terms of "erectile dysfunction" (AND) "stem cell therapy" (OR) "erectile dysfunction" (AND) "clinical trial of stem cell therapy" (OR) "stem cell therapy" (AND) "sexual dysfunction". The search was limited to English-language journals and full papers only. The initial search resulted in 450 articles, of which 90 relevant to our aims were included in the analysis. Results ED is a multifactorial disease. Current treatment options rely on pharmacotherapy as well as surgical options. Patients may have side effects or unsatisfactory results following the use of these treatment options. SCT may restore pathophysiological changes leading to ED rather than treating the symptoms. It has been evaluated in animal models and shown promising results in humans. Results confirm that SCT does improve erectile function in animals with different types of SC use. In humans, evidence showed promising results, but the trials were heterogeneous and limited mainly by a lack of randomization and the small sample size. Many challenges could limit future research in this field, including ethical dilemmas, regulation, patient recruitment, the cost of therapy, and the lack of a standardized SCT regimen. Repairing and possibly replacing diseased cells, tissue, or organs and eventually retrieving normal function should always be the goals of any therapy, and this can only be guaranteed by SCT. Conclusion SCT is a potential and successful treatment for ED, particularly in patients who are resistant to the classic therapy. SCT may promote nerve regeneration and vascular cell regeneration, not only symptomatic treatment.
Collapse
Affiliation(s)
| | - Hugo Bailly
- Department of Urology, Vivantes Klinikum, Berlin, Germany
| | - Fabian Klampke
- Department of Urology, Vivantes Klinikum, Berlin, Germany
| | - Johann Boaz
- Department of Urology, Royal Liverpool University Hospital, Liverpool, UK
| | | | - Ahmad Abou Yassine
- Department of Internal Medicine, Staten Island University Hospital, Staten Island, NY, USA
| | - Ian M. McElree
- Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mohamad Moussa
- Department of Urology, Lebanese University, Beirut, Lebanon
| |
Collapse
|
35
|
Bhat AA, Moglad E, Afzal M, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Ali H, Pant K, Singh TG, Dureja H, Singh SK, Dua K, Gupta G, Subramaniyan V. Therapeutic approaches targeting aging and cellular senescence in Huntington's disease. CNS Neurosci Ther 2024; 30:e70053. [PMID: 39428700 PMCID: PMC11491556 DOI: 10.1111/cns.70053] [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: 05/29/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disease that is manifested by a gradual loss of physical, cognitive, and mental abilities. As the disease advances, age has a major impact on the pathogenic signature of mutant huntingtin (mHTT) protein aggregation. This review aims to explore the intricate relationship between aging, mHTT toxicity, and cellular senescence in HD. Scientific data on the interplay between aging, mHTT, and cellular senescence in HD were collected from several academic databases, including PubMed, Google Scholar, Google, and ScienceDirect. The search terms employed were "AGING," "HUNTINGTON'S DISEASE," "MUTANT HUNTINGTIN," and "CELLULAR SENESCENCE." Additionally, to gather information on the molecular mechanisms and potential therapeutic targets, the search was extended to include relevant terms such as "DNA DAMAGE," "OXIDATIVE STRESS," and "AUTOPHAGY." According to research, aging leads to worsening HD pathophysiology through some processes. As a result of the mHTT accumulation, cellular senescence is promoted, which causes DNA damage, oxidative stress, decreased autophagy, and increased inflammatory responses. Pro-inflammatory cytokines and other substances are released by senescent cells, which may worsen the neuronal damage and the course of the disease. It has been shown that treatments directed at these pathways reduce some of the HD symptoms and enhance longevity in experimental animals, pointing to a new possibility of treating the condition. Through their amplification of the harmful effects of mHTT, aging and cellular senescence play crucial roles in the development of HD. Comprehending these interplays creates novel opportunities for therapeutic measures targeted at alleviating cellular aging and enhancing HD patients' quality of life.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of PharmacyPrince Sattam Bin Abdulaziz UniversityAl KharjSaudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy ProgramBatterjee Medical CollegeJeddahSaudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical SciencesUttaranchal UniversityDehradunIndia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of PharmacyUmm Al‐Qura UniversityMakkahSaudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of ScienceKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Sami I. Alzarea
- Department of Pharmacology, College of PharmacyJouf UniversitySakakaAl‐JoufSaudi Arabia
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Department of PharmacologyKyrgyz State Medical CollegeBishkekKyrgyzstan
| | - Kumud Pant
- Graphic Era (Deemed to be University), Dehradun, India
| | | | - Harish Dureja
- Department of Pharmaceutical SciencesMaharshi Dayanand UniversityRohtakIndia
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraPunjabIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of PharmacyChitkara UniversityRajpuraPunjabIndia
- Centre of Medical and Bio‐Allied Health Sciences ResearchAjman UniversityAjmanUnited Arab Emirates
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health SciencesMonash UniversityBandar SunwaySelangor Darul EhsanMalaysia
- Department of Medical SciencesSchool of Medical and Life Sciences Sunway UniversityBandar SunwaySelangor Darul EhsanMalaysia
| |
Collapse
|
36
|
Rodríguez-Eguren A, Bueno-Fernandez C, Gómez-Álvarez M, Francés-Herrero E, Pellicer A, Bellver J, Seli E, Cervelló I. Evolution of biotechnological advances and regenerative therapies for endometrial disorders: a systematic review. Hum Reprod Update 2024; 30:584-613. [PMID: 38796750 PMCID: PMC11369227 DOI: 10.1093/humupd/dmae013] [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: 12/07/2023] [Revised: 04/12/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND The establishment and maintenance of pregnancy depend on endometrial competence. Asherman syndrome (AS) and intrauterine adhesions (IUA), or endometrial atrophy (EA) and thin endometrium (TE), can either originate autonomously or arise as a result from conditions (i.e. endometritis or congenital hypoplasia), or medical interventions (e.g. surgeries, hormonal therapies, uterine curettage or radiotherapy). Affected patients may present an altered or inadequate endometrial lining that hinders embryo implantation and increases the risk of poor pregnancy outcomes and miscarriage. In humans, AS/IUA and EA/TE are mainly treated with surgeries or pharmacotherapy, however the reported efficacy of these therapeutic approaches remains unclear. Thus, novel regenerative techniques utilizing stem cells, growth factors, or tissue engineering have emerged to improve reproductive outcomes. OBJECTIVE AND RATIONALE This review comprehensively summarizes the methodologies and outcomes of emerging biotechnologies (cellular, acellular, and bioengineering approaches) to treat human endometrial pathologies. Regenerative therapies derived from human tissues or blood which were studied in preclinical models (in vitro and in vivo) and clinical trials are discussed. SEARCH METHODS A systematic search of full-text articles available in PubMed and Embase was conducted to identify original peer-reviewed studies published in English between January 2000 and September 2023. The search terms included: human, uterus, endometrium, Asherman syndrome, intrauterine adhesions, endometrial atrophy, thin endometrium, endometritis, congenital hypoplasia, curettage, radiotherapy, regenerative therapy, bioengineering, stem cells, vesicles, platelet-rich plasma, biomaterials, microfluidic, bioprinting, organoids, hydrogel, scaffold, sheet, miRNA, sildenafil, nitroglycerine, aspirin, growth hormone, progesterone, and estrogen. Preclinical and clinical studies on cellular, acellular, and bioengineering strategies to repair or regenerate the human endometrium were included. Additional studies were identified through manual searches. OUTCOMES From a total of 4366 records identified, 164 studies (3.8%) were included for systematic review. Due to heterogeneity in the study design and measured outcome parameters in both preclinical and clinical studies, the findings were evaluated qualitatively and quantitatively without meta-analysis. Groups using stem cell-based treatments for endometrial pathologies commonly employed mesenchymal stem cells (MSCs) derived from the human bone marrow or umbilical cord. Alternatively, acellular therapies based on platelet-rich plasma (PRP) or extracellular vesicles are gaining popularity. These are accompanied by the emergence of bioengineering strategies based on extracellular matrix (ECM)-derived hydrogels or synthetic biosimilars that sustain local delivery of cells and growth factors, reporting promising results. Combined therapies that target multiple aspects of tissue repair and regeneration remain in preclinical testing but have shown translational value. This review highlights the myriad of therapeutic material sources, administration methods, and carriers that have been tested. WIDER IMPLICATIONS Therapies that promote endometrial proliferation, vascular development, and tissue repair may help restore endometrial function and, ultimately, fertility. Based on the existing evidence, cost, accessibility, and availability of the therapies, we propose the development of triple-hit regenerative strategies, potentially combining high-yield MSCs (e.g. from bone marrow or umbilical cord) with acellular treatments (PRP), possibly integrated in ECM hydrogels. Advances in biotechnologies together with insights from preclinical models will pave the way for developing personalized treatment regimens for patients with infertility-causing endometrial disorders such as AS/IUA, EA/TE, and endometritis. REGISTRATION NUMBER https://osf.io/th8yf/.
Collapse
Affiliation(s)
- Adolfo Rodríguez-Eguren
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Clara Bueno-Fernandez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - María Gómez-Álvarez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Emilio Francés-Herrero
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Antonio Pellicer
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Rome, Rome, Italy
| | - José Bellver
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- Department of Paediatrics, Obstetrics and Gynecology, Faculty of Medicine, University of Valencia, Valencia, Spain
- IVIRMA Global Research Alliance, IVI Valencia, Valencia, Spain
| | - Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
- IVIRMA Global Research Alliance, IVIRMA New Jersey, Basking Ridge, NJ, USA
| | - Irene Cervelló
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| |
Collapse
|
37
|
Chugh RM, Bhanja P, Zitter R, Gunewardena S, Badkul R, Saha S. Modulation of β-Catenin is important to promote WNT expression in macrophages and mitigate intestinal injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.21.614209. [PMID: 39345507 PMCID: PMC11429945 DOI: 10.1101/2024.09.21.614209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Macrophages are the major source of WNT ligands. Macrophage-derived WNT is one of the most potent regenerative signals to mitigate intestinal injury. However, regulation of WNT expression in macrophages has not been studied. In the present study, we discovered that activation of canonical β-Catenin suppresses WNT expression in macrophages. Our CHIP-seq and validation study demonstrated the involvement of β-Catenin in the transcriptional regulation of WNT expression. Genetic and pharmacological approaches to de-stabilize/inactivate β-Catenin induce WNT expression in macrophages. Extracellular vesicles (EVs) are a major career of WNT ligands. Transfusion of EVs from pre-conditioned WNT-enriched macrophages demonstrated significant regenerative benefit over native macrophage-derived EVs to mitigate radiation-induced intestinal injury. Transfusion of WNT-enriched EVs also reduces DSS-induced colitis. Our study provides substantial evidence to consider that macrophage-targeted modulation of canonical WNT signaling to induce WNT expression followed by treatment with WNT-enriched EVs can be a lead therapy against intestinal injury.. SUMMARY Activation of β-Catenin suppresses WNT expression in macrophages. Macrophage-targeted pharmacological modulation of canonical WNT signaling followed by adoptive transfer mitigate radiation injury in intestine. EVs from these preconditioned macrophages mitigate chemical or radiation induced intestinal injury.
Collapse
|
38
|
Alquraisy A, Wilar G, Mohammed AFA, El-Rayyes A, Suhandi C, Wathoni N. A Comprehensive Review of Stem Cell Conditioned Media Role for Anti-Aging on Skin. Stem Cells Cloning 2024; 17:5-19. [PMID: 39310304 PMCID: PMC11416772 DOI: 10.2147/sccaa.s480437] [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: 05/29/2024] [Accepted: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Various studies have been widely conducted on conditioned medium for the development of anti-aging preparations, including the utilization of stem cells, which present a promising alternative solution. This narrative review aims to understand the latest developments in various conditioned medium stem cell applications for anti-aging on the skin. A search of the Scopus database yielded publications of interest. The research focused on articles published without restrictions on the year. After finding 68 articles in the search results, they moved on to the checking phase. Upon comprehensive literature review, 23 articles met the inclusion criteria, while 45 articles were deemed ineligible for participation in this research. The results of the review indicate that conditioned medium from various stem cells has demonstrated success in reducing risk factors for skin aging, as proven in various tests. The successful reduction of the risk of skin aging has been established in vitro, in vivo, and in clinical trials. Given the numerous studies on the progress of exploring and utilizing conditioned medium, it is expected to provide a solution to the problem of skin aging.
Collapse
Affiliation(s)
- Ayatulloh Alquraisy
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | | | - Ali El-Rayyes
- Department of Chemistry, College of Science, Northern Border University, Arar, Saudi Arabia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
39
|
Dogan NO, Suadiye E, Wrede P, Lazovic J, Dayan CB, Soon RH, Aghakhani A, Richter G, Sitti M. Immune Cell-Based Microrobots for Remote Magnetic Actuation, Antitumor Activity, and Medical Imaging. Adv Healthc Mater 2024; 13:e2400711. [PMID: 38885528 DOI: 10.1002/adhm.202400711] [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: 02/23/2024] [Revised: 05/17/2024] [Indexed: 06/20/2024]
Abstract
Translating medical microrobots into clinics requires tracking, localization, and performing assigned medical tasks at target locations, which can only happen when appropriate design, actuation mechanisms, and medical imaging systems are integrated into a single microrobot. Despite this, these parameters are not fully considered when designing macrophage-based microrobots. This study presents living macrophage-based microrobots that combine macrophages with magnetic Janus particles coated with FePt nanofilm for magnetic steering and medical imaging and bacterial lipopolysaccharides for stimulating macrophages in a tumor-killing state. The macrophage-based microrobots combine wireless magnetic actuation, tracking with medical imaging techniques, and antitumor abilities. These microrobots are imaged under magnetic resonance imaging and optoacoustic imaging in soft-tissue-mimicking phantoms and ex vivo conditions. Magnetic actuation and real-time imaging of microrobots are demonstrated under static and physiologically relevant flow conditions using optoacoustic imaging. Further, macrophage-based microrobots are magnetically steered toward urinary bladder tumor spheroids and imaged with a handheld optoacoustic device, where the microrobots significantly reduce the viability of tumor spheroids. The proposed approach demonstrates the proof-of-concept feasibility of integrating macrophage-based microrobots into clinic imaging modalities for cancer targeting and intervention, and can also be implemented for various other medical applications.
Collapse
Affiliation(s)
- Nihal Olcay Dogan
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
| | - Eylül Suadiye
- Materials Central Scientific Facility, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Paul Wrede
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
| | - Jelena Lazovic
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Cem Balda Dayan
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Ren Hao Soon
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
| | - Amirreza Aghakhani
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, 70569, Stuttgart, Germany
| | - Gunther Richter
- Materials Central Scientific Facility, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
- School of Medicine and College of Engineering, Koç University, Istanbul, 34450, Turkey
| |
Collapse
|
40
|
Artemova D, Vishnyakova P, Elchaninov A, Gantsova E, Sukhikh G, Fatkhudinov T. M1 macrophages as promising agents for cell therapy of endometriosis. Heliyon 2024; 10:e36340. [PMID: 39253270 PMCID: PMC11381802 DOI: 10.1016/j.heliyon.2024.e36340] [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: 03/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
Endometriosis is a chronic estrogen-dependent disease characterized by the presence of endometrial glands and stroma outside their normal anatomical location. While laparoscopic removal of foci remains the gold standard therapy, it has limited efficacy and certain risks. However, cell therapy using pro-inflammatory M1 macrophages presents a promising and minimally invasive alternative for treating endometriosis. This approach showcases the potential for innovative and effective treatments for this condition. This study aims to explore the anti-endometriosis properties of M1 macrophages. A reproducible syngeneic mouse model of endometriosis was utilized, revealing that formed foci are primarily composed of macrophages with an anti-inflammatory M2 phenotype rather than M1 macrophages. To investigate further, chemically reprogrammed M1 macrophages were labeled with the membrane fluorescent tag PKH26 and administered to animals with endometriosis. Therapy resulted in a decrease in the number and size of foci, accompanied by a shift in the phenotypic composition of peritoneal macrophages. Specifically, the content of M2 macrophages decreased while that of M1 macrophages increased, resembling the composition of healthy animals. Our study conclusively demonstrates the anti-endometriosis properties of M1 macrophages, providing a strong foundation for future research in the cell therapy of endometriosis.
Collapse
Affiliation(s)
- Daria Artemova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| | - Polina Vishnyakova
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Andrey Elchaninov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Elena Gantsova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 4 Oparina Street, 117997, Moscow, Russian Federation
| | - Timur Fatkhudinov
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution “Petrovsky National Research Centre of Surgery”, 3 Tsurupa Street, 117418, Moscow, Russian Federation
- Research Institute of Molecular and Cellular Medicine, RUDN University, 6 Miklukho-Maklaya Street, 117198, Moscow, Russian Federation
| |
Collapse
|
41
|
Sharma P, Maurya DK. Wharton's jelly mesenchymal stem cells: Future regenerative medicine for clinical applications in mitigation of radiation injury. World J Stem Cells 2024; 16:742-759. [PMID: 39086560 PMCID: PMC11287430 DOI: 10.4252/wjsc.v16.i7.742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/28/2024] [Accepted: 06/24/2024] [Indexed: 07/25/2024] Open
Abstract
Wharton's jelly mesenchymal stem cells (WJ-MSCs) are gaining significant attention in regenerative medicine for their potential to treat degenerative diseases and mitigate radiation injuries. WJ-MSCs are more naïve and have a better safety profile, making them suitable for both autologous and allogeneic transplantations. This review highlights the regenerative potential of WJ-MSCs and their clinical applications in mitigating various types of radiation injuries. In this review, we will also describe why WJ-MSCs will become one of the most probable stem cells for future regenerative medicine along with a balanced view on their strengths and weaknesses. Finally, the most updated literature related to both preclinical and clinical usage of WJ-MSCs for their potential application in the regeneration of tissues and organs will also be compiled.
Collapse
Affiliation(s)
- Prashasti Sharma
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India
| | - Dharmendra Kumar Maurya
- Life Sciences, Homi Bhabha National Institute, Mumbai 400094, Maharashtra, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, Maharashtra, India.
| |
Collapse
|
42
|
Semrau S. Neural Network-Based Filter Design for Compressive Raman Classification of Cells. J Chem Inf Model 2024; 64:5402-5412. [PMID: 38959402 PMCID: PMC11267571 DOI: 10.1021/acs.jcim.3c01856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/05/2024]
Abstract
Cell-based therapies are bound to revolutionize medicine, but significant technical hurdles must be overcome before wider adoption. In particular, nondestructive, label-free methods to characterize cells in real time are needed to optimize the production process and improve quality control. Raman spectroscopy, which provides a fingerprint of a cell's chemical composition, would be an ideal modality but is too slow for high-throughput applications. Compressive Raman techniques, which measure only linear combinations of Raman intensities, can be fast but require careful optimization to deliver high performance. Here, we develop a neural network model to identify optimal parameters for a compressive sensing scheme that reduces measurement time by 2 orders of magnitude. In a data set containing Raman spectra of three different cell types, it achieves up to 90% classification accuracy using only five linear combinations of Raman intensities. Our method thus unlocks the power of Raman spectroscopy for the characterization of cell products.
Collapse
|
43
|
Purcell E, Shah J, Powell C, Nguyen T, Zhou L, McDonald CA, Allison BJ, Malhotra A. Umbilical cord blood-derived therapy for preterm lung injury: a systematic review and meta-analysis. Stem Cells Transl Med 2024; 13:606-624. [PMID: 38819251 PMCID: PMC11227974 DOI: 10.1093/stcltm/szae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/14/2024] [Indexed: 06/01/2024] Open
Abstract
INTRODUCTION Lung injuries, such as bronchopulmonary dysplasia (BPD), remain a major complication of preterm birth, with limited therapeutic options. One potential emerging therapy is umbilical cord blood (UCB)-derived therapy. OBJECTIVES To systematically assess the safety and efficacy of UCB-derived therapy for preterm lung injury in preclinical and clinical studies. METHODS A systematic search of MEDLINE, Embase, CENTRAL, ClinicalTrials.gov, and WHO International Trials Registry Platform was performed. A meta-analysis was conducted with Review Manager (5.4.1) using a random effects model. Data was expressed as standardized mean difference (SMD) for preclinical data and pooled relative risk (RR) for clinical data, with 95% confidence intervals (CI). Potential effect modifiers were investigated via subgroup analysis. Certainty of evidence was assessed using the GRADE system. RESULTS Twenty-three preclinical studies and six clinical studies met eligibility criteria. Statistically significant improvements were seen across several preclinical outcomes, including alveolarization (SMD, 1.32, 95%CI [0.99, 1.65]), angiogenesis (SMD, 1.53, 95%CI [0.87, 2.18]), and anti-inflammatory cytokines (SMD, 1.68, 95%CI [1.03, 2.34]). In clinical studies, 103 preterm infants have received UCB-derived therapy for preterm lung injury and no significant difference was observed in the development of BPD (RR, 0.93, 95%CI [0.73, 1.18]). Across both preclinical and clinical studies, administration of UCB-derived therapy appeared safe. Certainty of evidence was assessed as "low." CONCLUSIONS Administration of UCB-derived therapy was associated with statistically significant improvements across several lung injury markers in preclinical studies. Early clinical studies demonstrated the administration of UCB-derived therapy as safe and feasible but lacked data regarding efficacy.
Collapse
Affiliation(s)
- Elisha Purcell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Jainam Shah
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Cameron Powell
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Timothy Nguyen
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
| | - Lindsay Zhou
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Beth J Allison
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Department of Obstetrics and Gynaecology, Monash University, VIC 3168, Melbourne, Australia
| | - Atul Malhotra
- Department of Paediatrics, Monash University, VIC 3168, Melbourne, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, VIC 3168, Melbourne, Australia
- Monash Newborn, Monash Children's Hospital, VIC 3168, Melbourne, Australia
| |
Collapse
|
44
|
Ye H, Zou X, Fang X. Advancing cell-based therapy in sepsis: An anesthesia outlook. Chin Med J (Engl) 2024; 137:1522-1534. [PMID: 38708689 PMCID: PMC11230747 DOI: 10.1097/cm9.0000000000003097] [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: 10/19/2023] [Indexed: 05/07/2024] Open
Abstract
ABSTRACT Sepsis poses a health challenge globally owing to markedly high rates of morbidity and mortality. Despite employing bundle therapy over two decades, approaches including transient organ supportive therapy and clinical trials focusing on signaling pathways have failed in effectively reversing multiple organ failure in patients with sepsis. Prompt and appropriate perioperative management for surgical patients with concurrent sepsis is urgent. Consequently, innovative therapies focusing on remedying organ injuries are necessitated. Cell therapy has emerged as a promising therapeutic avenue for repairing local damage to vital organs and restoring homeostasis during perioperative treatment for sepsis. Given the pivotal role of immune cell responses in the pathogenesis of sepsis, stem cell-based interventions that primarily modulate immune responses by interacting with multiple immune cells have progressed into clinical trials. The strides made in single-cell sequencing and gene-editing technologies have advanced the understanding of disease-specific immune responses in sepsis. Chimeric antigen receptor (CAR)-immune cell therapy offers an intriguing option for the treatment of sepsis. This review provides a concise overview of immune cell therapy, its current status, and the strides made in the context of sepsis research, discussing potential strategies for the management of patients with sepsis during perioperative stages.
Collapse
Affiliation(s)
- Hui Ye
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiaoyu Zou
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 312000, China
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
45
|
Lee NK, Chang JW. Manufacturing Cell and Gene Therapies: Challenges in Clinical Translation. Ann Lab Med 2024; 44:314-323. [PMID: 38361427 PMCID: PMC10961620 DOI: 10.3343/alm.2023.0382] [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: 09/26/2023] [Revised: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
The safety and efficacy of both cell and gene therapies have been demonstrated in numerous preclinical and clinical trials. Chimeric antigen receptor T (CAR-T) cell therapy, which leverages the technologies of both cell and gene therapies, has also shown great promise for treating various cancers. Advancements in pertinent fields have also highlighted challenges faced while manufacturing cell and gene therapy products. Potential problems and obstacles must be addressed to ease the clinical translation of individual therapies. Literature reviews of representative cell-based, gene-based, and cell-based gene therapies with regard to their general manufacturing processes, the challenges faced during manufacturing, and QC specifications are limited. We review the general manufacturing processes of cell and gene therapies, including those involving mesenchymal stem cells, viral vectors, and CAR-T cells. The complexities associated with the manufacturing processes and subsequent QC/validation processes may present challenges that could impede the clinical progression of the products. This article addresses these potential challenges. Further, we discuss the use of the manufacturing model and its impact on cell and gene therapy.
Collapse
Affiliation(s)
- Na Kyung Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jong Wook Chang
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
- Cell and Gene Therapy Institute (CGTI), Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Cell and Gene Therapy Institute, ENCell Co. Ltd., Seoul, Korea
| |
Collapse
|
46
|
Tian JS, Tay A. Progress on Electro-Enhancement of Cell Manufacturing. SMALL METHODS 2024; 8:e2301281. [PMID: 38059759 DOI: 10.1002/smtd.202301281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Indexed: 12/08/2023]
Abstract
With the long persistence of complex, chronic diseases in society, there is increasing motivation to develop cells as living medicine to treat diseases ranging from cancer to wounds. While cell therapies can significantly impact healthcare, the shortage of starter cells meant that considerable raw materials must be channeled solely for cell expansion, leading to expensive products with long manufacturing time which can prevent accessibility by patients who either cannot afford the treatment or have highly aggressive diseases and cannot wait that long. Over the last three decades, there has been increasing knowledge on the effects of electrical modulation on proliferation, but to the best of the knowledge, none of these studies went beyond how electro-control of cell proliferation may be extended to enhance industrial scale cell manufacturing. Here, this review is started by discussing the importance of maximizing cell yield during manufacturing before comparing strategies spanning biomolecular/chemical/physical to modulate cell proliferation. Next, the authors describe how factors governing invasive and non-invasive electrical stimulation (ES) including capacitive coupling electric field may be modified to boost cell manufacturing. This review concludes by describing what needs to be urgently performed to bridge the gap between academic investigation of ES to industrial applications.
Collapse
Affiliation(s)
- Johann Shane Tian
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
47
|
Salehi MS, Changiz Khani F, Ansari S, Mokhtari MJ, Dara M, Bayat M, Hooshmandi E, Ashjazadeh N, Borhani-Haghighi A, Ünal G, Pandamooz S. Hypoxic Preconditioning Prevents Oxidative Stress-Induced Cell Death in Human Hair Follicle Stem Cells. IRANIAN JOURNAL OF BIOTECHNOLOGY 2024; 22:e3888. [PMID: 39737203 PMCID: PMC11682525 DOI: 10.30498/ijb.2024.447077.3888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/19/2024] [Indexed: 01/01/2025]
Abstract
Objectives This study investigated the impact of hypoxic preconditioning on the survival and oxidative stress tolerance of nestin-expressing hair follicle stem cells (hHFSCs) and SH-SY5Y neuroblastoma cells, two crucial cell types for central nervous system therapies. The study also examined the relative expression of three key genes, HIF1α, BDNF, and VEGF following hypoxic preconditioning. Materials and Methods hHFSCs were isolated from human hair follicles, characterized, and subjected to hypoxia for up to 72 hours. SH-SY5Y cells were similarly preconditioned for up to 72 hours. Cell viability under hypoxic conditions and oxidative stress was assessed. The relative expression of key genes was evaluated using qRT-PCR. Results hHFSCs exhibited remarkable resilience to hypoxic conditions, while SH-SY5Y cells displayed lower tolerance. Hypoxic preconditioning improved the viability of both cell types under oxidative stress. HIF1α mRNA was significantly downregulated, and VEGF transcripts increased after preconditioning, suggesting adaptations to prolonged hypoxia. Conclusion Hypoxic preconditioning enhances the survival and oxidative stress resilience of hHFSCs and SH-SY5Y cells, offering potential benefits for central nervous system cell therapy. The differential responses observed emphasize the need for tailored preconditioning strategies for specific cell types. These findings underscore the importance of hypoxic preconditioning and warrant further research into the underlying mechanisms, bringing us closer to effective neurological disorder treatments.
Collapse
Affiliation(s)
- Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Sanaz Ansari
- Department of Biology, Zarghan Branch, Islamic Azad University, Zarghan, Iran
| | | | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Etrat Hooshmandi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nahid Ashjazadeh
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Gökhan Ünal
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
48
|
Tragoonlugkana P, Pruksapong C, Ontong P, Kamprom W, Supokawej A. Fibronectin and vitronectin alleviate adipose-derived stem cells senescence during long-term culture through the AKT/MDM2/P53 pathway. Sci Rep 2024; 14:14242. [PMID: 38902430 PMCID: PMC11189918 DOI: 10.1038/s41598-024-65339-z] [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: 02/19/2024] [Accepted: 06/19/2024] [Indexed: 06/22/2024] Open
Abstract
Cellular senescence plays a role in the development of aging-associated degenerative diseases. Cell therapy is recognized as a candidate treatment for degenerative diseases. To achieve the goal of cell therapy, the quality and good characteristics of cells are concerned. Cell expansion relies on two-dimensional culture, which leads to replicative senescence of expanded cells. This study aimed to investigate the effect of cell culture surface modification using fibronectin (FN) and vitronectin (VN) in adipose-derived stem cells (ADSCs) during long-term expansion. Our results showed that ADSCs cultured in FN and VN coatings significantly enhanced adhesion, proliferation, and slow progression of cellular senescence as indicated by lower SA-β-gal activities and decreased expression levels of genes including p16, p21, and p53. The upregulation of integrin α5 and αv genes influences phosphatidylinositol 4,5-bisphosphate 3-kinase (PI3K), and AKT proteins. FN and VN coatings upregulated AKT and MDM2 leading to p53 degradation. Additionally, MDM2 inhibition by Nutlin-3a markedly elevated p53 and p21 expression, increased cellular senescence, and induced the expression of inflammatory molecules including HMGB1 and IL-6. The understanding of FN and VN coating surface influencing ADSCs, especially senescence characteristics, offers a promising and practical point for the cultivation of ADSCs for future use in cell-based therapies.
Collapse
Affiliation(s)
- Patcharapa Tragoonlugkana
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Chatchai Pruksapong
- Department of Surgery, Phramongkutklao Hospital and Phramongkutklao College of Medicine, Bangkok, 10400, Thailand
| | - Pawared Ontong
- Department of Community Medical Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Witchayapon Kamprom
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
49
|
Weijler AM, Prickler L, Kainz V, Bergmann E, Bohle B, Regele H, Valenta R, Linhart B, Wekerle T. Adoptive Cell Therapy in Mice Sensitized to a Grass Pollen Allergen. Antibodies (Basel) 2024; 13:48. [PMID: 38920972 PMCID: PMC11200577 DOI: 10.3390/antib13020048] [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: 03/28/2024] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024] Open
Abstract
The proportion of patients with type I allergy in the world population has been increasing and with it the number of people suffering from allergic symptoms. Recently we showed that prophylactic cell therapy employing allergen-expressing bone marrow (BM) cells or splenic B cells induced allergen-specific tolerance in naïve mice. Here we investigated if cell therapy can modulate an established secondary allergen-specific immune response in pre-immunized mice. We sensitized mice against the grass pollen allergen Phl p 5 and an unrelated control allergen, Bet v 1, from birch pollen before the transfer of Phl p 5-expressing BM cells. Mice were conditioned with several combinations of low-dose irradiation, costimulation blockade, rapamycin and T cell-depleting anti-thymocyte globulin (ATG). Levels of allergen-specific IgE and IgG1 in serum after cell transfer were measured via ELISA and alterations in cellular responses were measured via an in vitro proliferation assay and transplantation of Phl p 5+ skin grafts. None of the tested treatment protocols impacted Phl p 5-specific antibody levels. Transient low-level chimerism of Phl p 5+ leukocytes as well as a markedly prolonged skin graft survival were observed in mice conditioned with high numbers of Phl p 5+ BMC or no sensitization events between the day of cell therapy and skin grafting. The data presented herein demonstrate that a pre-existing secondary allergen-specific immune response poses a substantial hurdle opposing tolerization through cell therapy and underscore the importance of prophylactic approaches for the prevention of IgE-mediated allergy.
Collapse
Affiliation(s)
- Anna Marianne Weijler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.M.W.); (L.P.); (V.K.); (E.B.)
| | - Lisa Prickler
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.M.W.); (L.P.); (V.K.); (E.B.)
| | - Verena Kainz
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.M.W.); (L.P.); (V.K.); (E.B.)
| | - Eva Bergmann
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.M.W.); (L.P.); (V.K.); (E.B.)
| | - Barbara Bohle
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (B.B.); (R.V.)
| | - Heinz Regele
- Clinical Institute of Pathology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (B.B.); (R.V.)
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- Institute of Immunology Federal Medical-Biological Agency (FMBA) of Russia, National Research Center (NRC), 119435 Moscow, Russia
- Laboratory of Immunopathology, Department of Clinical Immunology and Allergy, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Birgit Linhart
- Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria; (B.B.); (R.V.)
| | - Thomas Wekerle
- Division of Transplantation, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria; (A.M.W.); (L.P.); (V.K.); (E.B.)
| |
Collapse
|
50
|
Janes ME, Park KS, Gottlieb AP, Curreri A, Adebowale K, Kim J, Mitragotri S. Dendritic Cell Immune Modulation via Polyphenol Membrane Coatings. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28070-28079. [PMID: 38779939 DOI: 10.1021/acsami.4c01575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Cellular hitchhiking is an emerging strategy for the in vivo control of adoptively transferred immune cells. Hitchhiking approaches are primarily mediated by adhesion of nano and microparticles to the cell membrane, which conveys an ability to modulate transferred cells via local drug delivery. Although T cell therapies employing this strategy have progressed into the clinic, phagocytic cells including dendritic cells (DCs) are much more challenging to engineer. DC vaccines hold great potential for a spectrum of diseases, and the combination drug delivery is an attractive strategy to manipulate their function and overcome in vivo plasticity. However, DCs are not compatible with current hitchhiking approaches due to their broad phagocytic capacity. In this work, we developed and validated META (membrane engineering using tannic acid) to enable DC cellular hitchhiking for the first time. META employs the polyphenol tannic acid (TA) to facilitate supramolecular assembly of protein drug cargoes on the cell membrane, enabling the creation of cell surface-bound formulations for local drug delivery to carrier DCs. We optimized META formulations to incorporate and release protein cargoes with varying physical properties alone and in combination and to preserve DC viability and critical functions such as migration. We further show that META loaded with either a pro- or anti-inflammatory cargo can influence the carrier cell phenotype, thus demonstrating the flexibility of the approach for applications from cancer to autoimmune disease. Overall, this approach illustrates a new platform for the local control of phagocytic immune cells as a next step to advance DC therapies in the clinic.
Collapse
Affiliation(s)
- Morgan E Janes
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Alexander Curreri
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Kolade Adebowale
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Jayoung Kim
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| |
Collapse
|