1
|
Surico PL, Barone V, Singh RB, Coassin M, Blanco T, Dohlman TH, Basu S, Chauhan SK, Dana R, Di Zazzo A. Potential applications of mesenchymal stem cells in ocular surface immune-mediated disorders. Surv Ophthalmol 2025; 70:467-479. [PMID: 39097173 DOI: 10.1016/j.survophthal.2024.07.008] [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: 05/17/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We explore the interaction between corneal immunity and mesenchymal stem/stromal cells (MSCs) and their potential in treating corneal and ocular surface disorders. We outline the cornea's immune privilege mechanisms and the immunomodulatory substances involved. In this realm, MSCs are characterized by their immunomodulatory properties and regenerative potential, making them promising for therapeutic application. Therefore, we focus on the role of MSCs in immune-mediated corneal diseases such as dry eye disease, corneal transplantation rejection, limbal stem cell deficiency, and ocular graft-versus-host disease. Preclinical and clinical studies demonstrate MSCs' efficacy in promoting corneal healing and reducing inflammation in these conditions. Overall, we emphasize the potential of MSCs as innovative therapies in ophthalmology, offering promising solutions for managing various ocular surface pathologies.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Vincenzo Barone
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Marco Coassin
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas H Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sayan Basu
- Brien Holden Eye Research Centre (BHERC), L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Antonio Di Zazzo
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy.
| |
Collapse
|
2
|
Chen KY, Chan HC, Chan CM. Can Stem Cell Therapy Revolutionize Ocular Disease Treatment? A Critical Review of Preclinical and Clinical Advances. Stem Cell Rev Rep 2025:10.1007/s12015-025-10884-x. [PMID: 40266467 DOI: 10.1007/s12015-025-10884-x] [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: 04/08/2025] [Indexed: 04/24/2025]
Abstract
Stem cell therapy in regenerative medicine has a scope for treating ocular diseases. Stem cell therapy aims to repair damaged tissue and restore vision. The present review focuses on the advancements in stem cell therapies for ocular disorders, their mechanism of action, and clinical applications while addressing some outstanding challenges. Stem cells that include embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and retinal progenitor cells have regenerative potential for ocular repair. They differentiate into specialized ocular cell types, conduct neuroprotection, and modulate immune responses. It is emphasized in preclinical and clinical studies that stem cell therapy can treat corneal disorders such as limbal stem cell deficiency, retinal diseases like dry age macular degeneration and retinitis pigmentosa, and diabetic retinopathy. Various studies suggested that stem cells have considerable scope in glaucoma treatment by supporting retinal ganglion cell survival and optic nerve regeneration. Advanced approaches such as gene editing, organoid generation, and artificial intelligence enhance these therapies. Effective delivery to target areas, engraftment, orientation, and long-term survival of transplanted cells need optimization. Issues such as immune rejection and tumorigenicity must be addressed. This approach is further hindered by regulatory issues and overly complicated approval processes and trials. Ethical issues related to sourcing embryonic stem cells and patient consent complicate the issue. The cost of manufacturing stem cells and their accessibility are other factors posing potential barriers to widespread application. These regulatory, ethical, and economic issues must be tackled if stem cell treatments are to be made safe, accessible, and effective. Future studies will include refining therapeutic protocols, scaling manufacturing processes, and overcoming socio-economic barriers, eventually improving clinical outcomes.
Collapse
Affiliation(s)
- Kai-Yang Chen
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hoi-Chun Chan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chi-Ming Chan
- Department of Ophthalmology, Cardinal Tien Hospital, New Taipei City, Taiwan.
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.
| |
Collapse
|
3
|
Tan X, Zhang Z, Cao X, Qu L, Xiong Y, Li H, Wang Y, Chen Z, Shi C. Reprogramming of skin fibroblasts by 3D spheroid culture promotes peripheral nerve regeneration via the ID3/semaphorin7a pathway. Stem Cells Transl Med 2025; 14:szaf005. [PMID: 40213860 PMCID: PMC11986420 DOI: 10.1093/stcltm/szaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/31/2025] [Indexed: 04/14/2025] Open
Abstract
Peripheral nerve injury remains an intractable clinical issue with high morbidity, causing an excessive burden on the economy and society. Peripheral nerve tissue engineering combined with nerve conduits and supporting seed cells is considered a promising strategy for treating of long nerve defects. However, supporting seed cell sources that are easily accessible, capable of rapid expansion, and do not require genetic intervention are still urgently needed. This study intended to clarify whether the easily accessible and rapid expansion skin fibroblasts are the ideal supporting seed cells and can be reprogrammed into neural progenitor-like cells (NPCs) by forcing them to grow into a three-dimensional (3D) spheroid morphology. Results showed that 3D spheroid mouse dermal fibroblasts (MDFs) exhibited neural cell-like properties and could efficiently induce dorsal root ganglion neurons to extend the neurites. Transplantation of 3D spheroid MDFs significantly accelerated the regeneration of the sciatic nerve and improved the motor function of rats after transection compared to monolayer MDFs. Mechanism studies revealed that 3D spheroid culture significantly upregulated the expressions of the inhibitor of DNA binding 3 (ID3) and the hypoxia-inducible factor-1α (HIF-1α). The upregulation of the inhibitor of DNA binding 3 in 3D spheroid MDFs plays a critical role in acquiring NPC properties. Meanwhile, the upregulated ID3 and HIF-1α could synergistically upregulate semaphorin7a expression, which finally improved the extending of nerve axon in vitro and in vivo. This study may shed new light on treatments for peripheral nerve injury.
Collapse
Affiliation(s)
- Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Zhou Zhang
- Department of Ophthalmology, The Affiliated Hospital of Guizhou Medical University, Guizhou 550004, People’s Republic of China
| | - Xiaohui Cao
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Langfan Qu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Yinchun Xiong
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Huijuan Li
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Yu Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma and Chemical Poisoning, College of Preventive Medicine, Army Medical University, Chongqing 400038, People’s Republic of China
| |
Collapse
|
4
|
James MM, Zhou Y, Zhang M. Enhanced Differentiation of Human Neural Stem Cells into Cortical Neurons Using 3D Chitosan Scaffolds. ACS APPLIED BIO MATERIALS 2025; 8:2469-2481. [PMID: 40012088 DOI: 10.1021/acsabm.4c01927] [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: 02/28/2025]
Abstract
Human neural stem cells (hNSCs) have the potential to differentiate into various neural cell types, including cortical neurons, which are of particular interest for understanding and treating neurodegenerative diseases. However, traditional 2D culture methods are limited in their ability to accurately mimic the physiologically relevant microenvironment, leading to slow differentiation rates and low yields of mature neurons. In this study, we developed and optimized 3D chitosan scaffolds to promote the more efficient differentiation of hNSCs into cortical neurons. These scaffolds provide a tunable, biocompatible, and mechanically favorable environment, supporting enhanced cell-to-cell interactions and mimicking the extracellular matrix more effectively than 2D systems. The differentiation process was further accelerated by preseeding scaffolds with hNSCs, leading to increased expression of key cortical neuron markers, such as MAP2 and TUBB3, within a 14-day period. Compared to Geltrex-coated controls, the preseeded scaffolds demonstrated superior cell adhesion, viability, and differentiation efficiency, with significant upregulation of mature cortical neuron markers. Our findings suggest that chitosan-based 3D culture systems represent a promising platform for improving the differentiation of hNSCs, offering a faster and more reliable method to generate cortical neurons for neurodegenerative disease research and potential therapeutic applications.
Collapse
Affiliation(s)
- Matthew Michael James
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
5
|
Rajalekshmi R, Agrawal DK. Synergistic potential of stem cells and microfluidics in regenerative medicine. Mol Cell Biochem 2025; 480:1481-1493. [PMID: 39285093 PMCID: PMC11842489 DOI: 10.1007/s11010-024-05108-8] [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: 04/12/2024] [Accepted: 08/27/2024] [Indexed: 02/21/2025]
Abstract
Regenerative medicine has immense potential to revolutionize healthcare by using regenerative capabilities of stem cells. Microfluidics, a cutting-edge technology, offers precise control over cellular microenvironments. The integration of these two fields provides a deep understanding of stem cell behavior and enables the development of advanced therapeutic strategies. This critical review explores the use of microfluidic systems to culture and differentiate stem cells with precision. We examined the use of microfluidic platforms for controlled nutrient supply, mechanical stimuli, and real-time monitoring, providing an unprecedented level of detail in studying cellular responses. The convergence of stem cells and microfluidics holds immense promise for tissue repair, regeneration, and personalized medicine. It offers a unique opportunity to revolutionize the approach to regenerative medicine, facilitating the development of advanced therapeutic strategies and enhancing healthcare outcomes.
Collapse
Affiliation(s)
- Resmi Rajalekshmi
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
6
|
Fukutome A, Sakamoto T, Asawa Y, Riu D, Kawakami H, Hoshi K, Hikita A. Establishment of a mouse organ culture model of fetal cleft lip for the evaluation of adipose-derived stem cell therapy. Regen Ther 2025; 28:41-50. [PMID: 39687332 PMCID: PMC11647479 DOI: 10.1016/j.reth.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Cleft lip and cleft palate are congenital disorders resulting from abnormal facial development. Current treatments require multiple surgeries, which have risks of scar formation and facial deformities. Recently, fetal treatments utilizing "scarless healing" have gained attention, as early intervention shows potential to suppress scarring. In the field of regenerative medicine, mesenchymal stem cell therapies using cell sheets have advanced, by which promotion of tissue repair is expected. However, researches for fetal treatment using small animal models of cleft lip are challenging due to the high fetal mortality caused by surgical invasiveness. Although organ culture methods may offer an alternative approach, no organ culture system for fetal cleft lip research has been reported. Methods In this study, a cleft lip was surgically created on the upper left side lip of E15.5 mouse fetuses. These fetuses were cultured for four days using an organ culture system. Histological evaluation was performed to evaluate cell density, tissue morphology, and epithelialization. Additionally, adipose-derived stem cell (ADSC) sheets were transplanted two days after cleft lip creation to evaluate their effect on tissue repair. Results The histological analysis showed that cell density and tissue morphology were stably maintained in the four-day culture period. Epithelialization of the incision site was observed two days after surgery, confirming the completion of cleft formation. In the ADSC-transplanted group, epithelialization of the cleft site was observed, which indicates that the stem cell sheets contributed to tissue repair. Conclusion This research demonstrates the successful development of a cleft lip organ culture model and highlights the potential of ADSC sheets in promoting tissue repair. These findings provide a foundation for future regenerative medicine strategies in fetal cleft lip therapy.
Collapse
Affiliation(s)
- Ayane Fukutome
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tomoaki Sakamoto
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yukiyo Asawa
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Dan Riu
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroshi Kawakami
- Division of Dentistry and Oral Surgery, Mitsui Memorial Hospital, 1 Kanda Izumicho, Chiyoda-ku, Tokyo 101-8643, Japan
| | - Kazuto Hoshi
- Department of Oral and Maxillofacial Surgery, Dentistry and Orthodontics, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Atsuhiko Hikita
- Department of Tissue Engineering, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
7
|
Nissenbaum J, Segal E, Philip H, Cashman R, Golan‐Lev T, Reubinoff BE, Turjeman A, Yanuka O, Lezmi E, Kopper O, Benvenisty N. Predicting tumour resistance to paclitaxel and carboplatin utilising genome-wide screening in haploid human embryonic stem cells. Cell Prolif 2025; 58:e13771. [PMID: 39523021 PMCID: PMC11882768 DOI: 10.1111/cpr.13771] [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: 08/19/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Taxanes and platinum molecules, specifically paclitaxel and carboplatin, are widely used anticancer drugs that induce cell death and serve as first-line chemotherapy for various cancer types. Despite the efficient effect of both drugs on cancer cell proliferation, many tumours have innate resistance against paclitaxel and carboplatin, which leads to inefficient treatment and poor survival rates. Haploid human embryonic stem cells (hESCs) are a novel and robust platform for genetic screening. To gain a comprehensive view of genes that affect or regulate paclitaxel and carboplatin resistance, genome-wide loss-of-function screens in haploid hESCs were performed. Both paclitaxel and carboplatin screens have yielded selected plausible gene lists and pathways relevant to resistance prediction. The effects of mutations in selected genes on the resistance to the drugs were demonstrated. Based on the results, an algorithm that can predict resistance to paclitaxel or carboplatin was developed. Applying the algorithm to the DNA mutation profile of patients' tumours enabled the separation of sensitive versus resistant patients, thus, providing a prediction tool. As the anticancer drugs arsenal can offer alternatives in case of resistance to either paclitaxel or carboplatin, an early prediction can provide a significant advantage and should improve treatment. The algorithm assists this unmet need and helps predict whether a patient will respond to the treatment and may have an immediate clinically actionable application.
Collapse
Affiliation(s)
| | - Emanuel Segal
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and GynecologyHadassah Hebrew University Medical CenterJerusalemIsrael
| | | | | | - Tamar Golan‐Lev
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | - Benjamin E. Reubinoff
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and GynecologyHadassah Hebrew University Medical CenterJerusalemIsrael
| | - Adi Turjeman
- The Center for Genomic TechnologiesThe Hebrew UniversityJerusalemIsrael
| | - Ofra Yanuka
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| | | | | | - Nissim Benvenisty
- NewStem LTDJerusalemIsrael
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life SciencesThe Hebrew UniversityJerusalemIsrael
| |
Collapse
|
8
|
Lee MS, Lin ECY, Sivapatham A, Leiferman EM, Jiao H, Lu Y, Nemke BW, Leiferman M, Markel MD, Li WJ. Autologous iPSC- and MSC-derived chondrocyte implants for cartilage repair in a miniature pig model. Stem Cell Res Ther 2025; 16:86. [PMID: 39988676 PMCID: PMC11849328 DOI: 10.1186/s13287-025-04215-7] [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: 04/23/2024] [Accepted: 02/10/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cell (iPSC)-derived mesenchymal stem cells (iMSCs) have greater potential for generating chondrocytes without hypertrophic and fibrotic phenotypes compared to bone marrow-derived mesenchymal stem/stromal cells (BMSCs). However, there is a lack of research demonstrating the use of autologous iMSCs for repairing articular chondral lesions in large animal models. In this study, we aimed to evaluate the effectiveness of autologous miniature pig (minipig) iMSC-chondrocyte (iMSC-Ch)-laden implants in comparison to autologous BMSC-chondrocyte (BMSC-Ch)-laden implants for cartilage repair in porcine femoral condyles. METHODS iMSCs and BMSCs were seeded into fibrin glue/nanofiber constructs and cultured with chondrogenic induction media for 7 days before implantation. To assess the regenerative capacity of the cells, 19 skeletally mature Yucatan minipigs were randomly divided into microfracture control, acellular scaffold, iMSC, and BMSC subgroups. A cylindrical defect measuring 7 mm in diameter and 0.6 mm in depth was created on the articular cartilage surface without violating the subchondral bone. The defects were then left untreated or treated with acellular or cellular implants. RESULTS Both cellular implant-treated groups exhibited enhanced joint repair compared to the microfracture and acellular control groups. Immunofluorescence analysis yielded significant findings, showing that cartilage treated with iMSC-Ch implants exhibited higher expression of COL2A1 and minimal to no expression of COL1A1 and COL10A1, in contrast to the BMSC-Ch-treated group. This indicates that the iMSC-Ch implants generated more hyaline cartilage-like tissue compared to the BMSC-Ch implants. CONCLUSIONS Our findings contribute to filling the knowledge gap regarding the use of autologous iPSC derivatives for cartilage repair in a translational animal model. Moreover, these results highlight their potential as a safe and effective therapeutic strategy.
Collapse
Affiliation(s)
- Ming-Song Lee
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Eric Chang-Yi Lin
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Athillesh Sivapatham
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Ellen M Leiferman
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Hongli Jiao
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Yan Lu
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Brett W Nemke
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Matthew Leiferman
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Mark D Markel
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA
| | - Wan-Ju Li
- Musculoskeletal Biology and Regenerative Medicine Laboratory, Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, 53705, USA.
| |
Collapse
|
9
|
Zhou K, Qin Q, Lu J. Pathophysiological mechanisms of ARDS: a narrative review from molecular to organ-level perspectives. Respir Res 2025; 26:54. [PMID: 39948645 PMCID: PMC11827456 DOI: 10.1186/s12931-025-03137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) remains a life-threatening pulmonary condition with persistently high mortality rates despite significant advancements in supportive care. Its complex pathophysiology involves an intricate interplay of molecular and cellular processes, including cytokine storms, oxidative stress, programmed cell death, and disruption of the alveolar-capillary barrier. These mechanisms drive localized lung injury and contribute to systemic inflammatory response syndrome and multiple organ dysfunction syndrome. Unlike prior reviews that primarily focus on isolated mechanisms, this narrative review synthesizes the key pathophysiological processes of ARDS across molecular, cellular, tissue, and organ levels. MAIN BODY By integrating classical theories with recent research advancements, we provide a comprehensive analysis of how inflammatory mediators, metabolic reprogramming, oxidative stress, and immune dysregulation synergistically drive ARDS onset and progression. Furthermore, we critically evaluate current evidence-based therapeutic strategies, such as lung-protective ventilation and prone positioning, while exploring innovative therapies, including stem cell therapy, gene therapy, and immunotherapy. We emphasize the significance of ARDS subtypes and their inherent heterogeneity in guiding the development of personalized treatment strategies. CONCLUSIONS This narrative review provides fresh perspectives for future research, ultimately enhancing patient outcomes and optimizing management approaches in ARDS.
Collapse
Affiliation(s)
- Kaihuan Zhou
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Qianqian Qin
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China
| | - Junyu Lu
- Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, China.
| |
Collapse
|
10
|
Tan J, Chen J, Roxby D, Chooi WH, Nguyen TD, Ng SY, Han J, Chew SY. Using magnetic resonance relaxometry to evaluate the safety and quality of induced pluripotent stem cell-derived spinal cord progenitor cells. Stem Cell Res Ther 2024; 15:465. [PMID: 39639398 PMCID: PMC11622678 DOI: 10.1186/s13287-024-04070-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The emergence of induced pluripotent stem cells (iPSCs) offers a promising approach for replacing damaged neurons and glial cells, particularly in spinal cord injuries (SCI). Despite its merits, iPSC differentiation into spinal cord progenitor cells (SCPCs) is variable, necessitating reliable assessment of differentiation and validation of cell quality and safety. Phenotyping is often performed via label-based methods including immunofluorescent staining or flow cytometry analysis. These approaches are often expensive, laborious, time-consuming, destructive, and severely limits their use in large scale cell therapy manufacturing settings. On the other hand, cellular biophysical properties have demonstrated a strong correlation to cell state, quality and functionality and can be measured with ingenious label-free technologies in a rapid and non-destructive manner. METHOD In this study, we report the use of Magnetic Resonance Relaxometry (MRR), a rapid and label-free method that indicates iron levels based on its readout (T2). Briefly, we differentiated human iPSCs into SCPCs and compared key iPSC and SCPC cellular markers to their intracellular iron content (Fe3+) at different stages of the differentiation process. RESULTS With MRR, we found that intracellular iron of iPSCs and SCPCs were distinctively different allowing us to accurately reflect varying levels of residual undifferentiated iPSCs (i.e., OCT4+ cells) in any given population of SCPCs. MRR was also able to predict Day 10 SCPC OCT4 levels from Day 1 undifferentiated iPSC T2 values and identified poorly differentiated SCPCs with lower T2, indicative of lower neural progenitor (SOX1) and stem cell (Nestin) marker expression levels. Lastly, MRR was able to provide predictive indications for the extent of differentiation to Day 28 spinal cord motor neurons (ISL-1/SMI-32) based on the T2 values of Day 10 SCPCs. CONCLUSION MRR measurements of iPSCs and SCPCs has clearly indicated its capabilities to identify and quantify key phenotypes of iPSCs and SCPCs for end-point validation of safety and quality parameters. Thus, our technology provides a rapid label-free method to determine critical quality attributes in iPSC-derived progenies and is ideally suited as a quality control tool in cell therapy manufacturing.
Collapse
Affiliation(s)
- Jerome Tan
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- HealthTech @ NTU, Interdisciplinary Graduate Programme, Nanyang Technological University, Singapore, Singapore
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore
| | - Jiahui Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Daniel Roxby
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore
| | - Wai Hon Chooi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Shi Yan Ng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jongyoon Han
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, USA.
| | - Sing Yian Chew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- CAMP IRG, SMART Centre, CREATE, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
11
|
Hill M, Andrews-Pfannkoch C, Atherton E, Knudsen T, Trncic E, Marmorstein AD. Detection of Residual iPSCs Following Differentiation of iPSC-Derived Retinal Pigment Epithelial Cells. J Ocul Pharmacol Ther 2024; 40:680-687. [PMID: 39358867 PMCID: PMC11698679 DOI: 10.1089/jop.2024.0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
Purpose: The goal of this study was to develop a lot release assay for iPSC residuals following directed differentiation of iPSCs to retinal pigment epithelial (RPE) cells. Methods: RNA Sequencing (RNA Seq) of iPSCs and RPE derived from them was used to identify pluripotency markers downregulated in RPE cells. Quantitative real time PCR (qPCR) was then applied to assess iPSC residuals in iPSC-derived RPE. The limit of detection (LOD) of the assay was determined by performing spike-in assays with known quantities of iPSCs serially diluted into an RPE suspension. Results: ZSCAN10 and LIN28A were among 8 pluripotency markers identified by RNA Seq as downregulated in RPE. Based on copy number and expression of pseudogenes and lncRNAs ZSCAN10 and LIN28A were chosen for use in qPCR assays for residual iPSCs. Reverse transcription PCR indicated generally uniform expression of ZSCAN10 and LIN28A in 21 clones derived from 8 iPSC donors with no expression of either in RPE cells derived from 5 donor lines. Based on qPCR, ZSCAN10, and LIN28A expression in iPSCs was generally uniform. The LOD for ZSCAN10 and LIN28A in qPCR assays was determined using spike in assays of RPE derived from 2 iPSC lines. Analysis of ΔΔCt found the limit of detection to be <0.01% of cells, equivalent to <1 iPSC/10,000 RPE cells in both iPSC lines. Conclusions: qPCR for ZSCAN10 and LIN28A detects <1 in 10,000 residual iPSCs in a population of iPSC-derived RPE providing an adequate LOD of iPSC residuals for lot release testing.
Collapse
Affiliation(s)
- Matthew Hill
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Evan Atherton
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Travis Knudsen
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Emma Trncic
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
12
|
Wynne TM, Fritz VG, Simmons ZT, Zahed M, Seth A, Abbasi T, Reymundi MJ, Roballo KCS. Potential of Stem-Cell-Induced Peripheral Nerve Regeneration: From Animal Models to Clinical Trials. Life (Basel) 2024; 14:1536. [PMID: 39768245 PMCID: PMC11679741 DOI: 10.3390/life14121536] [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: 09/25/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Peripheral nerve injury has become an increasingly prevalent clinical concern, causing great morbidity in the community. Although there have been significant advancements in the treatment of peripheral nerve damage in recent years, the issue of long-term nerve regeneration remains. Furthermore, Wallerian degeneration has created an obstacle to long-term nerve regeneration. For this reason, there has been extensive research on the use of exogenous and endogenous stem cells as an adjunct or even primary treatment option for peripheral nerve injury. The plasticity and inducibility of stem cells make them an enticing option for initiating neuronal cell regrowth and optimal sensory and functional nerve regeneration. Peripheral nerve injury has a broad range of causative factors and etiologies. As such, unique stem cell-induced peripheral nerve treatments are being investigated to ameliorate the damage incited by all causes, including trauma, neuropathy, and systemic neurodegenerative diseases. This review is oriented to outline the potential role of stem cell therapies in peripheral nerve injury versus the current standards of care, compare the benefits and drawbacks of specific stem cell lines under investigation, and highlight the current models of stem cell therapy in the peripheral nervous system, with the ultimate goal of narrowing down and optimizing the role and scope of stem cell therapy in peripheral nerve injury.
Collapse
Affiliation(s)
- Taylor M. Wynne
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Virginia Grey Fritz
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Zachary T. Simmons
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Malek Zahed
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Ananya Seth
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Tamir Abbasi
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Michael J. Reymundi
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
| | - Kelly C. S. Roballo
- Department of Biomedical, Edward Via College of Osteopathic Medicine, Blacksburg, VA 24060, USA; (T.M.W.); (V.G.F.); (Z.T.S.); (M.Z.); (A.S.); (T.A.); (M.J.R.)
- Department of Biomedical Sciences and Pathobiology, Virginia Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
13
|
Krivec N, Couvreu de Deckersberg E, Lei Y, Al Delbany D, Regin M, Verhulst S, van Grunsven LA, Sermon K, Spits C. Gain of 1q confers an MDM4-driven growth advantage to undifferentiated and differentiating hESC while altering their differentiation capacity. Cell Death Dis 2024; 15:852. [PMID: 39572522 PMCID: PMC11582570 DOI: 10.1038/s41419-024-07236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
Gain of 1q is a highly recurrent chromosomal abnormality in human pluripotent stem cells. In this work, we show that gains of 1q impact the differentiation capacity to derivates of the three germ layers, leading to mis-specification to cranial placode and non-neural ectoderm during neuroectoderm differentiation. Also, we found a weaker expression of lineage-specific markers in hepatoblasts and cardiac progenitors. Competition assays show that the cells retain their selective advantage during differentiation, which is mediated by a higher expression of MDM4, a gene located in the common region of gain. MDM4 drives the winner phenotype of the mutant cells in both the undifferentiated and differentiating state by reducing the cells' sensitivity to DNA damage through decreased p53-mediated apoptosis. Finally, we found that cell density in culture plays a key role in promoting the competitive advantage of the cells by increasing DNA damage.
Collapse
Affiliation(s)
- Nuša Krivec
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Edouard Couvreu de Deckersberg
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Yingnan Lei
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Diana Al Delbany
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Marius Regin
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Stefaan Verhulst
- Liver Cell Biology Research Group, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Research Group, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Karen Sermon
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium
| | - Claudia Spits
- Research Group Genetics, Reproduction and Development, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090, Brussels, Belgium.
| |
Collapse
|
14
|
Alizadeh SD, Jahani S, Rukerd MRZ, Tabrizi R, Masoomi R, Banihashemian SZ, Tabatabaei MSHZ, Ghodsi Z, Pour-Rashidi A, Harrop J, Rahimi-Movaghar V. Human studies of the efficacy and safety of stem cells in the treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis. Stem Cell Res Ther 2024; 15:442. [PMID: 39563393 PMCID: PMC11577959 DOI: 10.1186/s13287-024-04033-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 10/30/2024] [Indexed: 11/21/2024] Open
Abstract
OBJECTIVE To assess the efficacy and safety of stem cell therapy in human studies for diabetic peripheral neuropathy (DPN). METHODS A comprehensive literature review was performed across multiple databases, including Ovid MEDLINE ALL, Embase via Ovid SP, Scopus, Web of Science Core Collection, and Cochrane CENTRAL, up to January 31, 2024. Keywords and controlled vocabularies related to diabetic neuropathy and stem cell therapy were used. Inclusion criteria encompassed all controlled trials examining stem cell therapy for DPN, excluding animal or in vitro studies, review papers, conference abstracts, and editor letters. Data extraction and risk of bias assessment were independently performed by multiple reviewers using standardized tools. RESULTS Out of 5431 initial entries, seven were included. Stem cell therapies included bone marrow-derived mononuclear cells and umbilical cord-derived mesenchymal stem cells, administered mainly via intramuscular transplantation. Meta-analysis indicated significant improvements in motor nerve conduction velocity (weighted mean differences (WMD): 2.2, 95% CI 1.6-2.8) and sensory nerve conduction velocity (WMD: 1.9, 95% CI 1.1-2.6). Vibration perception threshold and Toronto Clinical Scoring System scores decreased significantly (WMD: - 2.9, 95% CI - 4.0, - 1.8, and WMD: - 3.6, 95% CI - 5.0, - 2.2, respectively). Sensitivity analysis and subgroup analysis confirmed the robustness and specificity of these findings. The complications were pain and swelling at the injection sites, which disappeared in a few days. CONCLUSION Stem cell therapy shows significant promise in improving clinical outcomes for DPN, with evident benefits in nerve conduction and sensory parameters. Further research is needed to consolidate these findings and optimize therapeutic protocols.
Collapse
Affiliation(s)
- Seyed Danial Alizadeh
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Jahani
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rezaei Zadeh Rukerd
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Tabrizi
- Noncommunicable Diseases Research Center, Fasa University of Medical Science, Fasa, Iran
| | - Rasoul Masoomi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Zahra Ghodsi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Pour-Rashidi
- Department of Neurosurgery, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Founder of Neurosurgical Research Network, Universal Scientific Education and Research Network, Tehran, Iran
| | - James Harrop
- Department of Neurosurgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Universal Scientific Education and Research Network, Tehran, Iran.
| |
Collapse
|
15
|
Jiang Y, Harberts J, Assadi A, Chen Y, Spatz JP, Duan W, Nisbet DR, Voelcker NH, Elnathan R. The Roles of Micro- and Nanoscale Materials in Cell-Engineering Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410908. [PMID: 39401098 DOI: 10.1002/adma.202410908] [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: 07/26/2024] [Revised: 09/13/2024] [Indexed: 11/29/2024]
Abstract
Customizable manufacturing of ex vivo cell engineering is driven by the need for innovations in the biomedical field and holds substantial potential for addressing current therapeutic challenges; but it is still only in its infancy. Micro- and nanoscale-engineered materials are increasingly used to control core cell-level functions in cellular engineering. By reprogramming or redirecting targeted cells for extremely precise functions, these advanced materials offer new possibilities. This influences the modularity of cell reprogramming and reengineering, making these materials part of versatile and emerging technologies. Here, the roles of micro- and nanoscale materials in cell engineering are highlighted, demonstrating how they can be adaptively controlled to regulate cellular reprogramming and core cell-level functions, including differentiation, proliferation, adhesion, user-defined gene expression, and epigenetic changes. The current reprogramming routes used to achieve pluripotency from somatic cells and the significant potential of induced pluripotent stem cell technology for translational biomedical research are covered. Recent advances in nonviral intracellular delivery modalities for cell reprogramming and their constraints are evaluated. This paper focuses on emerging physical and combinatorial approaches of intracellular delivery for cell engineering, revealing the capabilities and limitations of these routes. It is showcased how these programmable materials are continually being explored as customizable tools for inducing biophysical stimulation. Harnessing the power of micro- and nanoscale-engineered materials will be a step change in the design of cell engineering, producing a suite of powerful tools for addressing potential future challenges in therapeutic cell engineering.
Collapse
Affiliation(s)
- Yuan Jiang
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - Jann Harberts
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Artin Assadi
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Yaping Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Zhejiang, 325000, China
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering (IMSE), Heidelberg University, 69120, Heidelberg, Germany
- Max Planck School Matter to Life, Max Planck Schools, 69120, Heidelberg, Germany
| | - Wei Duan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| | - David R Nisbet
- The Graeme Clark Institute, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, Parkville, VIC, 3010, Australia
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
| | - Roey Elnathan
- Faculty of Health, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, Victoria, 3216, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Victoria, Clayton, 3168, Australia
- Institute for Frontier Materials, Deakin University, Waurn Ponds, Victoria, 3216, Australia
| |
Collapse
|
16
|
Beaudreault CP, Wang R, Muh CR, Rosenberg A, Funari A, McGoldrick PE, Wolf SM, Sacknovitz A, Chung S. Overcoming Graft Rejection in Induced Pluripotent Stem Cell-Derived Inhibitory Interneurons for Drug-Resistant Epilepsy. Brain Sci 2024; 14:1027. [PMID: 39452039 PMCID: PMC11506040 DOI: 10.3390/brainsci14101027] [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: 08/26/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Cell-based therapies for drug-resistant epilepsy using induced pluripotent stem cell-derived inhibitory interneurons are now in early-phase clinical trials, building on findings from trials in Parkinson's disease (PD) and Huntington's disease (HD). Graft rejection and the need for immunosuppressive therapy post-transplantation pose potential barriers to more epilepsy patients becoming potential candidates for inhibitory interneurons transplantation surgery. OBJECTIVES The present literature review weighs the evidence for and against human leukocyte antigen (HLA)-mediated graft rejection in PD and HD and examines the potential advantages and drawbacks to five broad approaches to cell-based therapies, including autologous cell culture and transplantation, in vivo reprogramming of glial cells using viral vectors, allogeneic transplantation using off-the-shelf cell lines, transplantation using inhibitory interneurons cultured from HLA-matched cell lines, and the use of hypoimmunogenic-induced pluripotent stem cell-derived inhibitory interneurons. The impact of surgical technique and associated needle trauma on graft rejection is also discussed. METHODS Non-systematic literature review. RESULTS While cell-based therapies have enjoyed early successes in treating a host of central nervous system disorders, the immunologic reaction against surgical procedures and implanted materials has remained a major obstacle. CONCLUSIONS Adapting cell-based therapies using iPSC-derived inhibitory interneurons for epilepsy surgery will similarly require surmounting the challenge of immunogenicity.
Collapse
Affiliation(s)
- Cameron P. Beaudreault
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Richard Wang
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Carrie Rebecca Muh
- Department of Neurosurgery, Westchester Medical Center, Valhalla, NY 10595, USA
| | - Ashley Rosenberg
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
| | - Abigail Funari
- Department of Neurosurgery, SUNY Upstate Medical Center, Syracuse, NY 13210, USA
| | - Patty E. McGoldrick
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Division of Pediatric Neurology, Maria Fareri Children’s Hospital, Valhalla, NY 10595, USA
- Division of Pediatric Neurology, Boston Children’s Health Physicians, Hawthorne, NY 10532, USA
| | - Steven M. Wolf
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Division of Pediatric Neurology, Maria Fareri Children’s Hospital, Valhalla, NY 10595, USA
- Division of Pediatric Neurology, Boston Children’s Health Physicians, Hawthorne, NY 10532, USA
| | - Ariel Sacknovitz
- School of Medicine, New York Medical College, Valhalla, NY 10595, USA; (C.P.B.); (R.W.); (A.R.); (A.S.)
- Department of Neurosurgery, Westchester Medical Center, Valhalla, NY 10595, USA
| | - Sangmi Chung
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY 10595, USA
- Department of Neurosurgery, Brain Health Institute, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
17
|
de Oliveira Figueiredo EC, Bucolo C, Eandi CM. Therapeutic innovations for geographic atrophy: A promising horizon. Curr Opin Pharmacol 2024; 78:102484. [PMID: 39243634 DOI: 10.1016/j.coph.2024.102484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 05/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
This mini review spotlights the most promising treatments for geographic atrophy, the advanced form of age-related macular degeneration, often resulting in severe and irreversible vision loss. The pathophysiology is complex, and various therapeutic strategies, including anticomplement therapies, gene therapies, cell-based interventions, and artificial intelligence-driven diagnostics are discussed. Anticomplement therapies (antifactors C3 and C5) showed promise in reducing the inflammatory response and the progression of the atrophy. Gene therapies, targeting specific genetic mutations, are under development to correct underlying defects and potentially reverse disease progression. Cell-based therapies are gaining momentum, with early studies indicating encouraging results in the replacement of damaged retinal pigment epithelium cells.
Collapse
Affiliation(s)
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, Catania, Italy
| | - Chiara M Eandi
- Hôpital Ophtalmique Jules-Gonin, Fondation Asile des Aveugles, Lausanne, Switzerland; Department of Surgical Science, University of Torino, Torino, Italy.
| |
Collapse
|
18
|
Hussein KH, Ahmadzada B, Correa JC, Sultan A, Wilken S, Amiot B, Nyberg SL. Liver tissue engineering using decellularized scaffolds: Current progress, challenges, and opportunities. Bioact Mater 2024; 40:280-305. [PMID: 38973992 PMCID: PMC11226731 DOI: 10.1016/j.bioactmat.2024.06.001] [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: 04/03/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Liver transplantation represents the only definitive treatment for patients with end-stage liver disease. However, the shortage of liver donors provokes a dramatic gap between available grafts and patients on the waiting list. Whole liver bioengineering, an emerging field of tissue engineering, holds great potential to overcome this gap. This approach involves two main steps; the first is liver decellularization and the second is recellularization. Liver decellularization aims to remove cellular and nuclear materials from the organ, leaving behind extracellular matrices containing different structural proteins and growth factors while retaining both the vascular and biliary networks. Recellularization involves repopulating the decellularized liver with appropriate cells, theoretically from the recipient patient, to reconstruct the parenchyma, vascular tree, and biliary network. The aim of this review is to identify the major advances in decellularization and recellularization strategies and investigate obstacles for the clinical application of bioengineered liver, including immunogenicity of the designed liver extracellular matrices, the need for standardization of scaffold fabrication techniques, selection of suitable cell sources for parenchymal repopulation, vascular, and biliary tree reconstruction. In vivo transplantation models are also summarized for evaluating the functionality of bioengineered livers. Finally, the regulatory measures and future directions for confirming the safety and efficacy of bioengineered liver are also discussed. Addressing these challenges in whole liver bioengineering may offer new solutions to meet the demand for liver transplantation and improve patient outcomes.
Collapse
Affiliation(s)
- Kamal H. Hussein
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Anesthesiology, and Radiology, College of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Boyukkhanim Ahmadzada
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Julio Cisneros Correa
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Ahmer Sultan
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Silvana Wilken
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
19
|
Augustine R, Gezek M, Nikolopoulos VK, Buck PL, Bostanci NS, Camci-Unal G. Stem Cells in Bone Tissue Engineering: Progress, Promises and Challenges. Stem Cell Rev Rep 2024; 20:1692-1731. [PMID: 39028416 DOI: 10.1007/s12015-024-10738-y] [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: 05/17/2024] [Indexed: 07/20/2024]
Abstract
Bone defects from accidents, congenital conditions, and age-related diseases significantly impact quality of life. Recent advancements in bone tissue engineering (TE) involve biomaterial scaffolds, patient-derived cells, and bioactive agents, enabling functional bone regeneration. Stem cells, obtained from numerous sources including umbilical cord blood, adipose tissue, bone marrow, and dental pulp, hold immense potential in bone TE. Induced pluripotent stem cells and genetically modified stem cells can also be used. Proper manipulation of physical, chemical, and biological stimulation is crucial for their proliferation, maintenance, and differentiation. Stem cells contribute to osteogenesis, osteoinduction, angiogenesis, and mineralization, essential for bone regeneration. This review provides an overview of the latest developments in stem cell-based TE for repairing and regenerating defective bones.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Radiology, Stanford Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
| | - Mert Gezek
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | | | - Paige Lauren Buck
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Nazli Seray Bostanci
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA
- Biomedical Engineering and Biotechnology Graduate Program, University of Massachusetts, Lowell, MA, 01854, USA
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, MA, 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
20
|
Ore A, Angelastro JM, Giulivi C. Integrating Mitochondrial Biology into Innovative Cell Therapies for Neurodegenerative Diseases. Brain Sci 2024; 14:899. [PMID: 39335395 PMCID: PMC11429837 DOI: 10.3390/brainsci14090899] [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: 08/08/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The role of mitochondria in neurodegenerative diseases is crucial, and recent developments have highlighted its significance in cell therapy. Mitochondrial dysfunction has been implicated in various neurodegenerative disorders, including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis, and Huntington's diseases. Understanding the impact of mitochondrial biology on these conditions can provide valuable insights for developing targeted cell therapies. This mini-review refocuses on mitochondria and emphasizes the potential of therapies leveraging mesenchymal stem cells, embryonic stem cells, induced pluripotent stem cells, stem cell-derived secretions, and extracellular vesicles. Mesenchymal stem cell-mediated mitochondria transfer is highlighted for restoring mitochondrial health in cells with dysfunctional mitochondria. Additionally, attention is paid to gene-editing techniques such as mito-CRISPR, mitoTALENs, mito-ZNFs, and DdCBEs to ensure the safety and efficacy of stem cell treatments. Challenges and future directions are also discussed, including the possible tumorigenic effects of stem cells, off-target effects, disease targeting, immune rejection, and ethical issues.
Collapse
Affiliation(s)
- Adaleiz Ore
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- Department of Chemical Engineering, School of Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James M. Angelastro
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA; (A.O.); (J.M.A.)
- University of California Medical Investigations of Neurodevelopmental Disorders Institute (MIND Institute), University of California Health, Sacramento, CA 95817, USA
| |
Collapse
|
21
|
Liu S, Xu X, Omari-Siaw E, Yu J, Deng W. Progress of reprogramming astrocytes into neuron. Mol Cell Neurosci 2024; 130:103947. [PMID: 38862082 DOI: 10.1016/j.mcn.2024.103947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/13/2024] Open
Abstract
As the main players in the central nervous system (CNS), neurons dominate most life activities. However, after accidental trauma or neurodegenerative diseases, neurons are unable to regenerate themselves. The loss of this important role can seriously affect the quality of life of patients, ranging from movement disorders to disability and even death. There is no suitable treatment to prevent or reverse this process. Therefore, the regeneration of neurons after loss has been a major clinical problem and the key to treatment. Replacing the lost neurons by transdifferentiation of other cells is the only viable approach. Although much progress has been made in stem cell therapy, ethical issues, immune rejection, and limited cell sources still hinder its clinical application. In recent years, somatic cell reprogramming technology has brought a new dawn. Among them, astrocytes, as endogenously abundant cells homologous to neurons, have good potential and application value for reprogramming into neurons, having been reprogrammed into neurons in vitro and in vivo in a variety of ways.
Collapse
Affiliation(s)
- Sitong Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Science, Kumasi Technical University, PO Box 854, Kumasi, Ashanti, Ghana
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang, China; The International Institute on Natural Products and Stem Cells (iNPS), Zhenjiang, China; Key Lab for Drug Delivery & Tissue Regeneration, Zhenjiang, China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, China.
| |
Collapse
|
22
|
Intoh A, Watanabe-Susaki K, Kato T, Kiritani H, Kurisaki A. EPHA2 is a novel cell surface marker of OCT4-positive undifferentiated cells during the differentiation of mouse and human pluripotent stem cells. Stem Cells Transl Med 2024; 13:763-775. [PMID: 38811016 PMCID: PMC11328934 DOI: 10.1093/stcltm/szae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/14/2024] [Indexed: 05/31/2024] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) possess the intrinsic ability to differentiate into diverse cellular lineages, marking them as potent instruments in regenerative medicine. Nonetheless, the proclivity of these stem cells to generate teratomas post-transplantation presents a formidable obstacle to their therapeutic utility. In previous studies, we identified an array of cell surface proteins specifically expressed in the pluripotent state, as revealed through proteomic analysis. Here we focused on EPHA2, a protein found to be abundantly present on the surface of undifferentiated mouse ESCs and is diminished upon differentiation. Knock-down of Epha2 led to the spontaneous differentiation of mouse ESCs, underscoring a pivotal role of EPHA2 in maintaining an undifferentiated cell state. Further investigations revealed a strong correlation between EPHA2 and OCT4 expression during the differentiation of both mouse and human PSCs. Notably, removing EPHA2+ cells from mouse ESC-derived hepatic lineage reduced tumor formation after transplanting them into immune-deficient mice. Similarly, in human iPSCs, a larger proportion of EPHA2+ cells correlated with higher OCT4 expression, reflecting the pattern observed in mouse ESCs. Conclusively, EPHA2 emerges as a potential marker for selecting undifferentiated stem cells, providing a valuable method to decrease tumorigenesis risks after stem-cell transplantation in regenerative treatments.
Collapse
Affiliation(s)
- Atsushi Intoh
- Division of Biological Science, Nara Institute of Science and Technology, Nara, 630-0192, Japan
- Organ Development Research Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, 305-8560, Japan
| | - Kanako Watanabe-Susaki
- Organ Development Research Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, 305-8560, Japan
| | - Taku Kato
- Division of Biological Science, Nara Institute of Science and Technology, Nara, 630-0192, Japan
| | - Hibiki Kiritani
- Division of Biological Science, Nara Institute of Science and Technology, Nara, 630-0192, Japan
| | - Akira Kurisaki
- Division of Biological Science, Nara Institute of Science and Technology, Nara, 630-0192, Japan
- Organ Development Research Laboratory, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, 305-8560, Japan
| |
Collapse
|
23
|
Darsi SP, Baishya S, Nagati V, Bharani KK, Cheekatla SS, Darsi SK, Kamireddy AR, Barra RR, Devarasetti AK, Surampudi S, Singireddy JR, Kandula SK, Pasupulati AK. Safety assessment of rat embryonic fraction for in vivo regenerative therapy. Biol Open 2024; 13:bio060266. [PMID: 38984587 PMCID: PMC11360137 DOI: 10.1242/bio.060266] [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: 12/07/2023] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
Regenerative therapy is considered a novel option for treating various diseases, whereas a developing embryo is a prime source of molecules that help repair diseased tissue and organs. Organoid culture studies also confirmed the inherent biological functions of several embryonic factors. However, the in vivo safety and efficacy of embryonic protein fraction (EPF) were not validated. In this study, we investigated the effectiveness of EPF on healthy adult rats. We obtained embryos from Sprague-Dawley (SD) female rats of E14, E16, and E19 embryonic days and collected protein lysate. This lysate was administered intravenously into adult SD rats on sequential days. We collected blood and performed hematological and biochemical parameters of rats that received EPF. C-reactive protein levels, interleukin-6, blood glucose levels, serum creatinine, blood urea, total leucocyte counts, and % of neutrophils and lymphocytes were comparable between rats receiving EPF and saline. Histological examination of rats' tissues administered with EPF is devoid of abnormalities. Our study revealed that intravenous administration of EPF to healthy adult rats showed that EPF is non-immunogenic, non-inflammatory, non-tumorigenic, and safe for in vivo applications. Our analysis suggests that EPF or its components could be recommended for validating its therapeutic abilities in organ regenerative therapy.
Collapse
Affiliation(s)
- Sivarama Prasad Darsi
- Department of Biotechnology, School of Life Sciences, Gitam University, Visakhapatnam, AP, India530045
| | - Somorita Baishya
- Department of Biochemistry, University of Hyderabad, Hyderabad, TG, India500046
| | - Veerababu Nagati
- Department of Biochemistry, University of Hyderabad, Hyderabad, TG, India500046
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, P.V. Narasimha Rao University of Veterinary Sciences, Rajendra Nagar, TG, India500030
| | | | - Sujesh Kumar Darsi
- Department of General Medicine, ESI Corporation, Gunadala, Vijayawada, AP, India520004
| | - Adi Reddy Kamireddy
- Department of Internal Medicine, Banner Health Center, Maricopa, AZ, USA85138
| | - Ram Reddy Barra
- Department of Physiology, Apollo Institute of Medical Sciences and Research, Hyderabad, TG 500090, India
| | - Ashok Kumar Devarasetti
- Department of Veterinary Biochemistry, P.V. Narasimha Rao University of Veterinary Sciences, Mamnoor, Warangal, TG, India506166
| | - Sreedhar Surampudi
- Department of Biochemistry, Aware College of Medical Lab Technology, Bairamalguda, Hyderabad 500035, India
| | - Jayaram Reddy Singireddy
- Department of Urology, Hyderabad Kidney & Laparoscopic Centre, Malakpet, Hyderabad, TG 500036, India
| | - Siva Kumar Kandula
- Department of Biotechnology, School of Life Sciences, Gitam University, Visakhapatnam, AP, India530045
| | | |
Collapse
|
24
|
Zhang R, Chen Y, Feng Z, Cai B, Cheng Y, Du Y, Ou S, Chen H, Pan M, Liu H, Pei D, Cao S. Reprogramming human urine cells into intestinal organoids with long-term expansion ability and barrier function. Heliyon 2024; 10:e33736. [PMID: 39040281 PMCID: PMC11261862 DOI: 10.1016/j.heliyon.2024.e33736] [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: 12/21/2023] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
Generation of intestinal organoids from human somatic cells by reprogramming would enable intestinal regeneration, disease modeling, and drug screening in a personalized pattern. Here, we report a direct reprogramming protocol for the generation of human urine cells induced intestinal organoids (U-iIOs) under a defined medium. U-iIOs expressed multiple intestinal specific genes and showed resembling gene expression profiles to primary small intestines. U-iIOs can be stably long-term expanded and further differentiated into more mature intestinal lineage cells with high expression of metallothionein and cytochrome P450 (CYP450) genes. These specific molecular features of U-iIOs differ from human pluripotent stem cells derived intestinal organoids (P-iIOs) and intestinal immortalized cell lines. Furthermore, U-iIOs exhibit intestinal barriers indicated by blocking FITC-dextran permeation and uptaking of the specific substrate rhodamine 123. Our study provides a novel platform for patient-specific intestinal organoid generation, which may lead to precision treatment of intestinal diseases and facilitate drug discovery.
Collapse
Affiliation(s)
- Ruifang Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Yating Chen
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Ziyu Feng
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Baomei Cai
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Yiyi Cheng
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yunjing Du
- School of Biosciences & Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Sihua Ou
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Huan Chen
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Mengjie Pan
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - He Liu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou, China
| | - Shangtao Cao
- Key Laboratory of Biological Targeting Diagnosis, Therapy, and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Medical University, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Liu J, Kong G, Lu C, Wang J, Li W, Lv Z, Tong J, Liu Y, Xiong W, Li H, Fan J. IPSC-NSCs-derived exosomal let-7b-5p improves motor function after spinal cord Injury by modulating microglial/macrophage pyroptosis. J Nanobiotechnology 2024; 22:403. [PMID: 38982427 PMCID: PMC11232148 DOI: 10.1186/s12951-024-02697-w] [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: 02/14/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Following spinal cord injury (SCI), the inflammatory storm initiated by microglia/macrophages poses a significant impediment to the recovery process. Exosomes play a crucial role in the transport of miRNAs, facilitating essential cellular communication through the transfer of genetic material. However, the miRNAs from iPSC-NSCs-Exos and their potential mechanisms leading to repair after SCI remain unclear. This study aims to explore the role of iPSC-NSCs-Exos in microglia/macrophage pyroptosis and reveal their potential mechanisms. METHODS iPSC-NSCs-Exos were characterized and identified using transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot. A mouse SCI model and a series of in vivo and in vitro experiments were conducted to investigate the therapeutic effects of iPSC-NSCs-Exos. Subsequently, miRNA microarray analysis and rescue experiments were performed to confirm the role of miRNAs in iPSC-NSCs-Exos in SCI. Mechanistic studies were carried out using Western blot, luciferase activity assays, and RNA-ChIP. RESULTS Our findings revealed that iPSC-NSCs-derived exosomes inhibited microglia/macrophage pyroptosis at 7 days post-SCI, maintaining myelin integrity and promoting axonal growth, ultimately improving mice motor function. The miRNA microarray showed let-7b-5p to be highly enriched in iPSC-NSCs-Exos, and LRIG3 was identified as the target gene of let-7b-5p. Through a series of rescue experiments, we uncovered the connection between iPSC-NSCs and microglia/macrophages, revealing a novel target for treating SCI. CONCLUSION In conclusion, we discovered that iPSC-NSCs-derived exosomes can package and deliver let-7b-5p, regulating the expression of LRIG3 to ameliorate microglia/macrophage pyroptosis and enhance motor function in mice after SCI. This highlights the potential of combined therapy with iPSC-NSCs-Exos and let-7b-5p in promoting functional recovery and limiting inflammation following SCI.
Collapse
Affiliation(s)
- Jie Liu
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China
| | - Guang Kong
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chenlin Lu
- Department of Clinical Research Center, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Juan Wang
- Department of human anatomy, School of Basic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenbo Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China
| | - Zhengming Lv
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Jian Tong
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China
| | - Yuan Liu
- Songjiang Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wu Xiong
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China.
| | - Haijun Li
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China.
| | - Jin Fan
- Department of Orthopaedics, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Nanjing Medical University, 366 Taihu Road, Taizhou, Jiangsu, China.
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, China.
| |
Collapse
|
26
|
Chen KS, Koubek EJ, Sakowski SA, Feldman EL. Stem cell therapeutics and gene therapy for neurologic disorders. Neurotherapeutics 2024; 21:e00427. [PMID: 39096590 PMCID: PMC11345629 DOI: 10.1016/j.neurot.2024.e00427] [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: 02/26/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Rapid advances in biological knowledge and technological innovation have greatly advanced the fields of stem cell and gene therapies to combat a broad spectrum of neurologic disorders. Researchers are currently exploring a variety of stem cell types (e.g., embryonic, progenitor, induced pluripotent) and various transplantation strategies, each with its own advantages and drawbacks. Similarly, various gene modification techniques (zinc finger, TALENs, CRISPR-Cas9) are employed with various delivery vectors to modify underlying genetic contributors to neurologic disorders. While these two individual fields continue to blaze new trails, it is the combination of these technologies which enables genetically engineered stem cells and vastly increases investigational and therapeutic opportunities. The capability to culture and expand stem cells outside the body, along with their potential to correct genetic abnormalities in patient-derived cells or enhance cells with extra gene products, unleashes the full biological potential for innovative, multifaceted approaches to treat complex neurological disorders. In this review, we provide an overview of stem cell and gene therapies in the context of neurologic disorders, highlighting recent advances and current shortcomings, and discuss prospects for future therapies in clinical settings.
Collapse
Affiliation(s)
- Kevin S Chen
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily J Koubek
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Stacey A Sakowski
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
27
|
Bingnan W, Jiao T, Ghorbani A, Baghei S. Enhancing regenerative potential: A comprehensive review of stem cell transplantation for sports-related neuronal injuries, with a focus on spinal cord injuries and peripheral nervous system damage. Tissue Cell 2024; 88:102429. [PMID: 38833939 DOI: 10.1016/j.tice.2024.102429] [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: 01/24/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Neuronal injuries, as one of the consequences of sports-related incidents, exert a profound influence on the athletes' future, potentially leading to complete immobility and impeding their athletic pursuits. In cases of severe damage inflicted upon the spinal cord (SC) and peripheral nervous systems (PNS), the regenerative process is notably compromised, rendering it essentially inefficient. Among the pivotal therapeutic approaches for the enhancement and prevention of secondary SC injuries (SCI), stem cell transplantation (SCT) stands out prominently. Stem cells, whether directly involved in replacement and reconstruction or indirectly through modification and secretion of crucial bioenvironmental factors, engage in the intricate process of tissue regeneration. Stem cells, through the secretion of neurotrophic factors (NTFs) (aiming to modulate the immune system), reduction of inflammation, axonal growth stimulation, and myelin formation, endeavor to facilitate the regeneration of damaged SC tissue. The fundamental challenges of this approach encompass the proper selection of suitable stem cell candidates for transplantation and the establishment of an appropriate microenvironment conducive to SC repair. In this article, an attempt has been made to explore sports-related injuries, particularly SCI, to comprehensively review innovative methods for treating SCI, and to address the existing challenges. Additionally, some of the stem cells used in neural injuries and the process of their utilization have been discussed.
Collapse
Affiliation(s)
- Wang Bingnan
- Department of P.E, Central South University, Changsha 410083, China
| | - Tong Jiao
- The High School Attached to Hunan Normal University Bocai Experimental Middle School,Changsha 410208, China.
| | - A Ghorbani
- Biotechnology Department, Islamic Azad University, Isfahan, Iran
| | - Sh Baghei
- Biotechnology Department, Islamic Azad University, Isfahan, Iran.
| |
Collapse
|
28
|
Lin YC, Ku CC, Wuputra K, Liu CJ, Wu DC, Satou M, Mitsui Y, Saito S, Yokoyama KK. Possible Strategies to Reduce the Tumorigenic Risk of Reprogrammed Normal and Cancer Cells. Int J Mol Sci 2024; 25:5177. [PMID: 38791215 PMCID: PMC11120835 DOI: 10.3390/ijms25105177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
The reprogramming of somatic cells to pluripotent stem cells has immense potential for use in regenerating or redeveloping tissues for transplantation, and the future application of this method is one of the most important research topics in regenerative medicine. These cells are generated from normal cells, adult stem cells, or neoplastic cancer cells. They express embryonic stem cell markers, such as OCT4, SOX2, and NANOG, and can differentiate into all tissue types in adults, both in vitro and in vivo. However, tumorigenicity, immunogenicity, and heterogeneity of cell populations may hamper the use of this method in medical therapeutics. The risk of cancer formation is dependent on mutations of these stemness genes during the transformation of pluripotent stem cells to cancer cells and on the alteration of the microenvironments of stem cell niches at genetic and epigenetic levels. Recent reports have shown that the generation of induced pluripotent stem cells (iPSCs) derived from human fibroblasts could be induced using chemicals, which is a safe, easy, and clinical-grade manufacturing strategy for modifying the cell fate of human cells required for regeneration therapies. This strategy is one of the future routes for the clinical application of reprogramming therapy. Therefore, this review highlights the recent progress in research focused on decreasing the tumorigenic risk of iPSCs or iPSC-derived organoids and increasing the safety of iPSC cell preparation and their application for therapeutic benefits.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cha-Chien Ku
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- Waseda Research Institute for Science and Engineering, Waseda University, Tokyo 169-8555, Japan
| | - Chung-Jung Liu
- Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (D.-C.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Deng-Chyang Wu
- Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (D.-C.W.)
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Maki Satou
- Research Institute, Horus Co., Ltd., Iruma 358-0032, Saitama, Japan; (M.S.); (Y.M.)
| | - Yukio Mitsui
- Research Institute, Horus Co., Ltd., Iruma 358-0032, Saitama, Japan; (M.S.); (Y.M.)
| | - Shigeo Saito
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.)
- Research Institute, Horus Co., Ltd., Iruma 358-0032, Saitama, Japan; (M.S.); (Y.M.)
- Saito Laboratory of Cell Technology, Yaita 329-1571, Tochigi, Japan
| | - Kazunari K. Yokoyama
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Graduate Institute of Medicine, Department of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-J.L.); (D.-C.W.)
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| |
Collapse
|
29
|
Zhou Z, Li Y, Wu S, Liu T, Jiang J. Host-microbiota interactions in collagen-induced arthritis rats treated with human umbilical cord mesenchymal stem cell exosome and ginsenoside Rh2. Biomed Pharmacother 2024; 174:116515. [PMID: 38569276 DOI: 10.1016/j.biopha.2024.116515] [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: 01/21/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Mesenchymal stem cell exosome (MSCs-exo) is a class of products secreted by mesenchymal stem cells (MSCs) that contain various biologically active substances. MSCs-exo is a promising alternative to MSCs due to their lower immunogenicity and lack of ethical constraints. Ginsenoside Rh2 (Rh2) is a hydrolyzed component of the primary active substance of ginsenosides. Rh2 has a variety of pharmacological functions, including anti-inflammatory, anti-tumor, and antioxidant. Studies have demonstrated that gut microbiota and metabolites are critical in developing rheumatoid arthritis (RA). In this study, we constructed a collagen-induced arthritis (CIA) model in rats. We used MSCs-exo combined with Rh2 to treat CIA rats. To observe the effect of MSCs-exo combined with Rh2 on joint inflammation, rat feces were collected for 16 rRNA amplicon sequencing and untargeted metabolomics analysis. The results showed that the arthritis index score and joint swelling of CIA rats treated with MSCs-exo in combination with Rh2 were significantly lower than those of the model and MSCs-exo alone groups. MSCs-exo and Rh2 significantly ameliorated the disturbed gut microbiota in CIA rats. The regulation of Candidatus_Saccharibacteria and Clostridium_XlVb regulation may be the most critical. Rh2 enhanced the therapeutic effect of MSCs-exo compared with the MSCs-exo -alone group. Furthermore, significant changes in gut metabolites were observed in the CIA rat group, and these differentially altered metabolites may act as messengers for host-microbiota interactions. These differential metabolites were enriched into relevant critical metabolic pathways, revealing possible pathways for host-microbiota interactions.
Collapse
Affiliation(s)
- Zhongsheng Zhou
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shuhui Wu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China; Yibin Jilin University Research Institute, Jilin University, Yibin, Sichuan, China.
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
30
|
Ren R, Jiang J, Li X, Zhang G. Research progress of autoimmune diseases based on induced pluripotent stem cells. Front Immunol 2024; 15:1349138. [PMID: 38720903 PMCID: PMC11076788 DOI: 10.3389/fimmu.2024.1349138] [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: 12/04/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.
Collapse
Affiliation(s)
| | | | | | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| |
Collapse
|
31
|
Sheikhlary S, Lopez DH, Moghimi S, Sun B. Recent Findings on Therapeutic Cancer Vaccines: An Updated Review. Biomolecules 2024; 14:503. [PMID: 38672519 PMCID: PMC11048403 DOI: 10.3390/biom14040503] [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: 04/06/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer remains one of the global leading causes of death and various vaccines have been developed over the years against it, including cell-based, nucleic acid-based, and viral-based cancer vaccines. Although many vaccines have been effective in in vivo and clinical studies and some have been FDA-approved, there are major limitations to overcome: (1) developing one universal vaccine for a specific cancer is difficult, as tumors with different antigens are different for different individuals, (2) the tumor antigens may be similar to the body's own antigens, and (3) there is the possibility of cancer recurrence. Therefore, developing personalized cancer vaccines with the ability to distinguish between the tumor and the body's antigens is indispensable. This paper provides a comprehensive review of different types of cancer vaccines and highlights important factors necessary for developing efficient cancer vaccines. Moreover, the application of other technologies in cancer therapy is discussed. Finally, several insights and conclusions are presented, such as the possibility of using cold plasma and cancer stem cells in developing future cancer vaccines, to tackle the major limitations in the cancer vaccine developmental process.
Collapse
Affiliation(s)
- Sara Sheikhlary
- Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | - David Humberto Lopez
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Sophia Moghimi
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| | - Bo Sun
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA; (D.H.L.); (S.M.)
| |
Collapse
|
32
|
Sun T, Li M, Liu Q, Yu A, Cheng K, Ma J, Murphy S, McNutt PM, Zhang Y. Insights into optimizing exosome therapies for acute skin wound healing and other tissue repair. Front Med 2024; 18:258-284. [PMID: 38216854 PMCID: PMC11283324 DOI: 10.1007/s11684-023-1031-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/15/2023] [Indexed: 01/14/2024]
Abstract
Exosome therapy holds great promise as a novel approach to improve acute skin wound healing. This review provides a comprehensive overview of the current understanding of exosome biology and its potential applications in acute skin wound healing and beyond. Exosomes, small extracellular vesicles secreted by various stem cells, have emerged as potent mediators of intercellular communication and tissue repair. One advantage of exosome therapy is its ability to avoid potential risks associated with stem cell therapy, such as immune rejection or stem cells differentiating into unwanted cell types. However, further research is necessary to optimize exosome therapy, not only in the areas of exosome isolation, characterization, and engineering, but also in determining the optimal dose, timing, administration, and frequency of exosome therapy. Thus, optimization of exosome therapy is critical for the development of more effective and safer exosome-based therapies for acute skin wound healing and other diseases induced by cancer, ischemia, or inflammation. This review provides valuable insights into the potential of exosome therapy and highlights the need for further research to optimize exosome therapy for clinical use.
Collapse
Affiliation(s)
- Tianjing Sun
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China
| | - Mo Li
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China
| | - Qi Liu
- Department of Nephrology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China.
| | - Anyong Yu
- Department of Emergency, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, China.
| | - Kun Cheng
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, 64108, USA
| | - Jianxing Ma
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Sean Murphy
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Patrick Michael McNutt
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA
| | - Yuanyuan Zhang
- Wake Forest Institute of Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
33
|
Hersh AM, Weber-Levine C, Jiang K, Theodore N. Spinal Cord Injury: Emerging Technologies. Neurosurg Clin N Am 2024; 35:243-251. [PMID: 38423740 DOI: 10.1016/j.nec.2023.10.001] [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: 03/02/2024]
Abstract
The mainstay of treatment for spinal cord injury includes decompressive laminectomy and elevation of mean arterial pressure. However, outcomes often remain poor. Extensive research and ongoing clinical trials seek to design new treatment options for spinal cord injury, including stem cell therapy, scaffolds, brain-spine interfaces, exoskeletons, epidural electrical stimulation, ultrasound, and cerebrospinal fluid drainage. Some of these treatments are targeted at the initial acute window of injury, during which secondary damage occurs; others are designed to help patients living with chronic injuries.
Collapse
Affiliation(s)
- Andrew M Hersh
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA. https://twitter.com/AndrewMHersh
| | - Carly Weber-Levine
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA
| | - Kelly Jiang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA. https://twitter.com/kellyjjiang
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Meyer 7-113, Baltimore, MD 21287, USA; Orthopaedic Surgery & Biomedical Engineering, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
34
|
Surico PL, Scarabosio A, Miotti G, Grando M, Salati C, Parodi PC, Spadea L, Zeppieri M. Unlocking the versatile potential: Adipose-derived mesenchymal stem cells in ocular surface reconstruction and oculoplastics. World J Stem Cells 2024; 16:89-101. [PMID: 38455097 PMCID: PMC10915950 DOI: 10.4252/wjsc.v16.i2.89] [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: 12/07/2023] [Revised: 01/06/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
This review comprehensively explores the versatile potential of mesenchymal stem cells (MSCs) with a specific focus on adipose-derived MSCs. Ophthalmic and oculoplastic surgery, encompassing diverse procedures for ocular and periocular enhancement, demands advanced solutions for tissue restoration, functional and aesthetic refinement, and aging. Investigating immunomodulatory, regenerative, and healing capacities of MSCs, this review underscores the potential use of adipose-derived MSCs as a cost-effective alternative from bench to bedside, addressing common unmet needs in the field of reconstructive and regenerative surgery.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA 02114, United States
- Department of Ophthalmology, Campus Bio-Medico University, Rome 00128, Italy
| | - Anna Scarabosio
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Giovanni Miotti
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Martina Grando
- Department of Internal Medicine, Azienda Sanitaria Friuli Occidentale, San Vito al Tagliamento 33078, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Pier Camillo Parodi
- Department of Plastic Surgery, University Hospital of Udine, Udine 33100, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00142, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy.
| |
Collapse
|
35
|
Hadzimustafic N, D’Elia A, Shamoun V, Haykal S. Human-Induced Pluripotent Stem Cells in Plastic and Reconstructive Surgery. Int J Mol Sci 2024; 25:1863. [PMID: 38339142 PMCID: PMC10855589 DOI: 10.3390/ijms25031863] [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: 12/12/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
A hallmark of plastic and reconstructive surgery is restoring form and function. Historically, tissue procured from healthy portions of a patient's body has been used to fill defects, but this is limited by tissue availability. Human-induced pluripotent stem cells (hiPSCs) are stem cells derived from the de-differentiation of mature somatic cells. hiPSCs are of particular interest in plastic surgery as they have the capacity to be re-differentiated into more mature cells, and cultured to grow tissues. This review aims to evaluate the applications of hiPSCs in the plastic surgery context, with a focus on recent advances and limitations. The use of hiPSCs and non-human iPSCs has been researched in the context of skin, nerve, vasculature, skeletal muscle, cartilage, and bone regeneration. hiPSCs offer a future for regenerated autologous skin grafts, flaps comprised of various tissue types, and whole functional units such as the face and limbs. Also, they can be used to model diseases affecting tissues of interest in plastic surgery, such as skin cancers, epidermolysis bullosa, and scleroderma. Tumorigenicity, immunogenicity and pragmatism still pose significant limitations. Further research is required to identify appropriate somatic origin and induction techniques to harness the epigenetic memory of hiPSCs or identify methods to manipulate epigenetic memory.
Collapse
Affiliation(s)
- Nina Hadzimustafic
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Andrew D’Elia
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Valentina Shamoun
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; (N.H.); (A.D.); (V.S.)
| | - Siba Haykal
- Department of Plastic and Reconstructive Surgery, University Health Network, Toronto, ON M5G 2C4, Canada
| |
Collapse
|
36
|
Frost ER, Gilchrist RB. Making human eggs in a dish: are we close? Trends Biotechnol 2024; 42:168-178. [PMID: 37625913 DOI: 10.1016/j.tibtech.2023.07.007] [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: 05/17/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023]
Abstract
In the space of 50 years, we have seen incredible achievements in human reproductive medicine. With these leaps forward, it is no wonder that there is a major interest in women's reproductive health research, including extension of reproductive lifespan. Substantial effort is currently being made to address this challenge, including from the commercial sector. In vitro gametogenesis (IVG) in mice is a spectacular breakthrough and has the potential to offer hope to women with intractable infertility. However, with such lofty goals, some reflection may be called for: mastering all of the techniques required for complete and safe IVG in women is likely to be extraordinarily difficult.
Collapse
Affiliation(s)
- Emily R Frost
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Robert B Gilchrist
- Fertility & Research Centre, Discipline of Women's Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
37
|
Liu DH, Tseng HC, Lee MS, Chiou GY, Wang CT, Lin YY, Lai WY, Liu YH, Wang CY, Lee CY, Kao CL, Chen CF, Chien Y. Overcoming the challenges of scalable iPSC generation in translation medicine. J Chin Med Assoc 2024; 87:163-170. [PMID: 38132887 DOI: 10.1097/jcma.0000000000001046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The potential of induced pluripotent stem cells (iPSCs) in revolutionizing regenerative medicine cannot be overstated. iPSCs offer a profound opportunity for therapies involving cell replacement, disease modeling, and cell transplantation. However, the widespread application of iPSC cellular therapy faces hurdles, including the imperative to regulate iPSC differentiation rigorously and the inherent genetic disparities among individuals. To address these challenges, the concept of iPSC super donors emerges, holding exceptional genetic attributes and advantageous traits. These super donors serve as a wellspring of standardized, high-quality cell sources, mitigating inter-individual variations and augmenting the efficacy of therapy. METHODS In pursuit of this goal, our study embarked on the establishment of iPSC cell lines specifically sourced from donors possessing the HLA type (A33:03-B58:01-DRB1*03:01). The reprogramming process was meticulously executed, resulting in the successful generation of iPSC lines from these carefully selected donors. Subsequently, an extensive characterization was conducted to comprehensively understand the features and attributes of these iPSC lines. RESULTS The outcomes of our research were highly promising. The reprogramming efforts culminated in the generation of iPSC lines from donors with the specified HLA type. These iPSC lines displayed a range of distinctive characteristics that were thoroughly examined and documented. This successful generation of iPSC lines from super donors possessing advantageous genetic traits represents a significant stride towards the realization of their potential in therapeutic applications. CONCLUSION In summary, our study marks a crucial milestone in the realm of regenerative medicine. The establishment of iPSC lines from super donors with specific HLA types signifies a paradigm shift in addressing challenges related to iPSC cellular therapy. The standardized and high-quality cell sources derived from these super donors hold immense potential for various therapeutic applications. As we move forward, these findings provide a solid foundation for further research and development, ultimately propelling the field of regenerative medicine toward new horizons of efficacy and accessibility.
Collapse
Affiliation(s)
- Ding-Hao Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Huan-Chin Tseng
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Meng-Shiue Lee
- Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Guang-Yuh Chiou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Chin-Tien Wang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
- Division of Clinical Research, Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yi-Ying Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Wei-Yi Lai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Yu-Hao Liu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chien-Ying Wang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Exercise and Health Sciences, University of Taipei, Taipei, Taiwan, ROC
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Division of Trauma, Department of Emergency Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Chen-Yi Lee
- Department of Electronics Engineering and Institute of Electronics, National Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Chung-Lan Kao
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan, ROC
| | - Cheng-Fong Chen
- Department of Exercise and Health Sciences, University of Taipei, Taipei, Taiwan, ROC
- Department of Orthopaedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Surgery, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yueh Chien
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| |
Collapse
|
38
|
Cross-Najafi AA, Farag K, Chen AM, Smith LJ, Zhang W, Li P, Ekser B. The Long Road to Develop Custom-built Livers: Current Status of 3D Liver Bioprinting. Transplantation 2024; 108:357-368. [PMID: 37322580 PMCID: PMC10724374 DOI: 10.1097/tp.0000000000004668] [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] [Indexed: 06/17/2023]
Abstract
Although liver transplantation is the gold-standard therapy for end-stage liver disease, the shortage of suitable organs results in only 25% of waitlisted patients undergoing transplants. Three-dimensional (3D) bioprinting is an emerging technology and a potential solution for personalized medicine applications. This review highlights existing 3D bioprinting technologies of liver tissues, current anatomical and physiological limitations to 3D bioprinting of a whole liver, and recent progress bringing this innovation closer to clinical use. We reviewed updated literature across multiple facets in 3D bioprinting, comparing laser, inkjet, and extrusion-based printing modalities, scaffolded versus scaffold-free systems, development of an oxygenated bioreactor, and challenges in establishing long-term viability of hepatic parenchyma and incorporating structurally and functionally robust vasculature and biliary systems. Advancements in liver organoid models have also increased their complexity and utility for liver disease modeling, pharmacologic testing, and regenerative medicine. Recent developments in 3D bioprinting techniques have improved the speed, anatomical, and physiological accuracy, and viability of 3D-bioprinted liver tissues. Optimization focusing on 3D bioprinting of the vascular system and bile duct has improved both the structural and functional accuracy of these models, which will be critical in the successful expansion of 3D-bioprinted liver tissues toward transplantable organs. With further dedicated research, patients with end-stage liver disease may soon be recipients of customized 3D-bioprinted livers, reducing or eliminating the need for immunosuppressive regimens.
Collapse
Affiliation(s)
- Arthur A. Cross-Najafi
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristine Farag
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Angela M. Chen
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lester J. Smith
- Department of Radiology and Imaging Sciences, Indiana University of School of Medicine, Indianapolis, IN, USA
- 3D Bioprinting Core, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wenjun Zhang
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ping Li
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
39
|
Sun N, Sun Y, Xing Y, Xu L, Chen Z, Qing L, Wu P, Tang J. Knowledge mapping and research trends of stem cell in wound healing: A bibliometric analysis. Int Wound J 2024; 21:e14587. [PMCID: PMC10830390 DOI: 10.1111/iwj.14587] [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: 10/31/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2025] Open
Abstract
Wound nonhealing is a common and difficult problem in clinic. Stem cells are pluripotent cells, and their undifferentiated and self‐replicating characteristics have attracted much attention in the regenerative medicine‐related researches. New treatment approaches might result from an understanding of the function of stem cells in wound healing. Using bibliometric techniques, this study proposed to analyse the research status, hotspots, and research trends in stem cell and wound healing. By using the Web of Science Core Collection (WoSCC), we conducted an in‐depth review of publications on stem cells in wound healing from 1999 to 2023. We used scientometric analysis methods to examine annual trends, institutions, countries, journals, authors, keywords, co‐occurrence references and their closed relationship, revealing present hotspots and potential future advancements in this field. We analysed 19 728 English studies and discovered a consistent rise in annual publications. The United States and China were the two countries with the most publications. The most three influential institutions in the field were Shanghai Jiao Tong University, Sun Yat‐sen University, and University of Pittsburgh. International Journal of Molecular Sciences and Biomaterials were considered the most influential journals in this field. International Journal of Molecular Sciences had the most publications, and the most quantity of citations and the highest H‐index were found in Biomaterials . The dual‐map overlay revealed that publications in Molecular/Biology/Genetics and Health/Nursing/Medicine co‐cited journals received the majority of the citations for studies from Molecular/Biology/Immunology and Medicine/Medical/Clinical. In terms of publication production and influence, Fu X stood out among the authors, and Pittenger MF took the top spot in co‐citations. According to the keywords from the analysis, future research should concentrate on the mechanisms through which stem cells promote wound healing. We conducted a thorough analysis of the general information, knowledge base and research hotspots in the field of stem cells and wound healing from 1999 to 2023 by using the VOSviewer, CiteSpace, and other bibliometric analysis tools. It not only provided valuable insights for scholars, but also served as a reliable reference that drives further development in the field and stimulates the interest of researchers.
Collapse
Affiliation(s)
- Nianzhe Sun
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Yu Sun
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Yixuan Xing
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
- Department of EmergencyXiangya Hospital, Central South UniversityChangshaChina
| | - Laiyu Xu
- Department of OrthopedicsThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Zijie Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Liming Qing
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Panfeng Wu
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| | - Juyu Tang
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
40
|
Krivec N, Ghosh MS, Spits C. Gains of 20q11.21 in human pluripotent stem cells: Insights from cancer research. Stem Cell Reports 2024; 19:11-27. [PMID: 38157850 PMCID: PMC10828824 DOI: 10.1016/j.stemcr.2023.11.013] [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: 09/08/2023] [Revised: 11/28/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
The genetic abnormalities observed in hPSC cultures worldwide have been suggested to pose an important hurdle in their safe use in regenerative medicine due to the possibility of oncogenic transformation by mutant cells in the patient posttransplantation. One of the best-characterized genetic lesions in hPSCs is the gain of 20q11.21, found in 20% of hPSC lines worldwide, and strikingly, also amplified in 20% of human cancers. In this review, we have curated the existing knowledge on the incidence of this mutation in hPSCs and cancer, explored the significance of chromosome 20q11.21 amplification in cancer progression, and reviewed the oncogenic role of the genes in the smallest common region of gain, to shed light on the significance of this mutation in hPSC-based cell therapy. Lastly, we discuss the state-of-the-art strategies devised to detect aneuploidies in hPSC cultures, avoid genetic changes in vitro cultures of hPSCs, and strategies to eliminate genetically abnormal cells from culture.
Collapse
Affiliation(s)
- Nuša Krivec
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Manjusha S Ghosh
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Claudia Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
41
|
Kashima M, Komura R, Sato Y, Hashimoto C, Hirata H. A resource of single-cell gene expression profiles in a planarian Dugesia japonica. Dev Growth Differ 2024; 66:43-55. [PMID: 37779230 DOI: 10.1111/dgd.12893] [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: 02/21/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023]
Abstract
The freshwater planarian Dugesia japonica maintains an abundant heterogeneous cell population called neoblasts, which include adult pluripotent stem cells. Thus, it is an excellent model organism for stem cell and regeneration research. Recently, many single-cell RNA sequencing (scRNA-seq) databases of several model organisms, including other planarian species, have become publicly available; these are powerful and useful resources to search for gene expression in various tissues and cells. However, the only scRNA-seq dataset for D. japonica has been limited by the number of genes detected. Herein, we collected D. japonica cells, and conducted an scRNA-seq analysis. A novel, automatic, iterative cell clustering strategy produced a dataset of 3,404 cells, which could be classified into 63 cell types based on gene expression profiles. We introduced two examples for utilizing the scRNA-seq dataset in this study using D. japonica. First, the dataset provided results consistent with previous studies as well as novel functionally relevant insights, that is, the expression of DjMTA and DjP2X-A genes in neoblasts that give rise to differentiated cells. Second, we conducted an integrative analysis of the scRNA-seq dataset and time-course bulk RNA-seq of irradiated animals, demonstrating that the dataset can help interpret differentially expressed genes captured via bulk RNA-seq. Using the R package "Seurat" and GSE223927, researchers can easily access and utilize this dataset.
Collapse
Affiliation(s)
- Makoto Kashima
- College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
- Department of Molecular Biology, Faculty of Science, Toho University, Funabashi, Japan
| | - Rei Komura
- College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| | - Yuki Sato
- JT Biohistory Research Hall, Takatsuki, Japan
| | - Chikara Hashimoto
- JT Biohistory Research Hall, Takatsuki, Japan
- Department of Biological Sciences, Graduate School of Science, Osaka University, Toyonaka, Japan
| | - Hiromi Hirata
- College of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| |
Collapse
|
42
|
Guo T, Wang J, Pang M, Liu W, Zhang X, Fan A, Liu H, Liu Q, Wei T, Li C, Zhao X, Lu Y. Reprogramming and multi-lineage transdifferentiation attenuate the tumorigenicity of colorectal cancer cells. J Biol Chem 2024; 300:105534. [PMID: 38072050 PMCID: PMC10801221 DOI: 10.1016/j.jbc.2023.105534] [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: 05/29/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 01/02/2024] Open
Abstract
Significant advances have been made in reprogramming various somatic cells into induced pluripotent stem cells (iPSCs) and in multi-lineage differentiation (transdifferentiation) into different tissues. These manipulable transdifferentiating techniques may be applied in cancer therapy. Limited works have been reported that cancer cell malignancy can be switched to benign phenotypes through reprogramming techniques. Here, we reported that two colorectal cancer (CRC) cell lines (DLD1, HT29) could be reprogrammed into iPSCs (D-iPSCs, H-iPSCs). D- and H-iPSCs showed reduced tumorigenesis. Furthermore, we successfully induced D- and H-iPSCs differentiation into terminally differentiated cell types such as cardiomyocyte, neuron, and adipocyte-like cells. Impressively, the differentiated cells exhibited further attenuated tumorigenesis in vitro and in vivo. RNA-Seq further indicated that epigenetic changes occurred after reprogramming and transdifferentiation that caused reduced tumorigenicity. Overall, our study indicated that CRC cells can be reprogrammed and further differentiated into terminally differentiated lineages with attenuation of their malignancy in vitro and in vivo. The current work sheds light on a potential multi-lineage differentiation therapeutic strategy for colorectal cancer.
Collapse
Affiliation(s)
- Tongtong Guo
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juan Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Maogui Pang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wanning Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaohui Zhang
- Department of Gastroenterology, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ahui Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hengtao Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Qianqian Liu
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Tianying Wei
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China
| | - Cunxi Li
- Jiaen Genetics Laboratory, Beijing Jiaen Hospital, Beijing, China; Cytogenetics Laboratory, Beijing Institute of Human Genetics and Reproduction Medicine, Beijing, China.
| | - Xiaodi Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yuanyuan Lu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
43
|
Zhu W, Li M, Zou J, Zhang D, Fang M, Sun Y, Li C, Tang M, Wang Y, Zhou Q, Zhao T, Li W, Hu Z, Hu B. Induction of local immunosuppression in allogeneic cell transplantation by cell-type-specific expression of PD-L1 and CTLA4Ig. Stem Cell Reports 2023; 18:2344-2355. [PMID: 37995700 PMCID: PMC10724073 DOI: 10.1016/j.stemcr.2023.10.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/25/2023] Open
Abstract
Immune rejection has long hindered allogeneic cell transplantation therapy. Current genetic modification approaches, including direct targeting of major histocompatibility complex or constitutive expression of immune inhibitory molecules, exhibit drawbacks such as severe adverse effects or elevated tumorigenesis risks. To overcome these limitations, we introduce an innovative approach to induce cell-type-specific immune tolerance in differentiated cells. By engineering human embryonic stem cells, we ensure the exclusive production of the immune inhibitory molecules PD-L1/CTLA4Ig in differentiated cells. Using this strategy, we generated hepatocyte-like cells expressing PD-L1 and CTLA4Ig, which effectively induced local immunotolerance. This approach was evaluated in a humanized mouse model that mimics the human immune system dynamics. We thus demonstrate a robust and selective induction of immunotolerance specific to hepatocytes, improving graft survival without observed tumorigenesis. This precise immune tolerance strategy holds great promise for advancing the development of stem cell-based therapeutics in regenerative medicine.
Collapse
Affiliation(s)
- Wenliang Zhu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Mengqi Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jun Zou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Minghui Fang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China
| | - Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Can Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Zheng Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital of Jilin University, Jilin, China; National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, Jilin 130061, China.
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
44
|
Hasanzadeh A, Ebadati A, Dastanpour L, Aref AR, Sahandi Zangabad P, Kalbasi A, Dai X, Mehta G, Ghasemi A, Fatahi Y, Joshi S, Hamblin MR, Karimi M. Applications of Innovation Technologies for Personalized Cancer Medicine: Stem Cells and Gene-Editing Tools. ACS Pharmacol Transl Sci 2023; 6:1758-1779. [PMID: 38093832 PMCID: PMC10714436 DOI: 10.1021/acsptsci.3c00102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 02/16/2024]
Abstract
Personalized medicine is a new approach toward safer and even cheaper treatments with minimal side effects and toxicity. Planning a therapy based on individual properties causes an effective result in a patient's treatment, especially in a complex disease such as cancer. The benefits of personalized medicine include not only early diagnosis with high accuracy but also a more appropriate and effective therapeutic approach based on the unique clinical, genetic, and epigenetic features and biomarker profiles of a specific patient's disease. In order to achieve personalized cancer therapy, understanding cancer biology plays an important role. One of the crucial applications of personalized medicine that has gained consideration more recently due to its capability in developing disease therapy is related to the field of stem cells. We review various applications of pluripotent, somatic, and cancer stem cells in personalized medicine, including targeted cancer therapy, cancer modeling, diagnostics, and drug screening. CRISPR-Cas gene-editing technology is then discussed as a state-of-the-art biotechnological advance with substantial impacts on medical and therapeutic applications. As part of this section, the role of CRISPR-Cas genome editing in recent cancer studies is reviewed as a further example of personalized medicine application.
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Arefeh Ebadati
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Lida Dastanpour
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Advances
Nanobiotechnology and Nanomedicine Research Group (ANNRG), Iran University of Medical Sciences, Tehran 14535, Iran
| | - Amir R. Aref
- Department
of Medical Oncology and Belfer Center for Applied Cancer Science, Dana Farber Cancer Institute, Boston, Massachusetts 02115, United States
| | - Parham Sahandi Zangabad
- Monash
Institute of Pharmaceutical Sciences, Department of Pharmacy and Pharmaceutical
Sciences, Monash University, Parkville, Melbourne, Victoria 3052, Australia
| | - Alireza Kalbasi
- Department
of Medical Oncology, Dana-Farber Cancer
Institute, Boston, Massachusetts 02115, United States
| | - Xiaofeng Dai
- School of
Biotechnology, Jiangnan University, Wuxi 214122, China
- National
Engineering Laboratory for Cereal Fermentation Technology, Jiangnan University, Wuxi 214122, China
- Jiangsu Provincial
Research Center for Bioactive Product Processing Technology, Jiangnan University, Wuxi 214122, China
| | - Geeta Mehta
- Department
of Biomedical Engineering, University of
Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Materials Science and Engineering, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Macromolecular
Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
- Rogel Cancer
Center, University of Michigan, Ann Arbor, Michigan 48109, United States
- Precision
Health, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Amir Ghasemi
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Department
of Materials Science and Engineering, Sharif
University of Technology, Tehran 14588, Iran
| | - Yousef Fatahi
- Nanotechnology
Research Centre, Faculty of Pharmacy, Tehran
University of Medical Sciences, Tehran 14166, Iran
- Department
of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14166, Iran
- Universal
Scientific Education and Research Network (USERN), Tehran 14166, Iran
| | - Suhasini Joshi
- Chemical
Biology Program, Memorial Sloan Kettering
Cancer Center, New York, New York 10065, United States
| | - Michael R. Hamblin
- Laser Research
Centre, Faculty of Health Science, University
of Johannesburg, Doornfontein 2028, South Africa
- Radiation
Biology Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
| | - Mahdi Karimi
- Cellular
and Molecular Research Center, Iran University
of Medical Sciences, Tehran 14535, Iran
- Department
of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
- Oncopathology
Research Center, Iran University of Medical
Sciences, Tehran 14535, Iran
- Research
Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 14166, Iran
- Applied
Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 14166, Iran
| |
Collapse
|
45
|
Wong DCS, Harvey JP, Jurkute N, Thomasy SM, Moosajee M, Yu-Wai-Man P, Gilhooley MJ. OPA1 Dominant Optic Atrophy: Pathogenesis and Therapeutic Targets. J Neuroophthalmol 2023; 43:464-474. [PMID: 37974363 PMCID: PMC10645107 DOI: 10.1097/wno.0000000000001830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Affiliation(s)
- David C. S. Wong
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Joshua P. Harvey
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Neringa Jurkute
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Sara M. Thomasy
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Mariya Moosajee
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Patrick Yu-Wai-Man
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| | - Michael J. Gilhooley
- Department of Clinical Neurosciences (DCSW, PY-W-M), John van Geest Center for Brain Repair, University of Cambridge, Cambridge, United Kingdom; Cambridge Eye Unit (DCSW, PY-W-M), Addenbrooke's Hospital, Cambridge, United Kingdom; UCL Institute of Ophthalmology (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Moorfields Eye Hospital NHS Foundation Trust (JPH, NJ, MM, PY-W-M, MJG), London, United Kingdom; Department of Ophthalmology and Vision Science (SMT), School of Medicine, U.C. Davis, Sacramento, California; Department of Surgical and Radiological Sciences (SMT), School of Veterinary Medicine, U.C. Davis, California; Great Ormond Street Hospital (MM), London, United Kingdom; and The Francis Crick Institute (MM), London, United Kingdom
| |
Collapse
|
46
|
Li X, Fan R, Xiang J, Yuan Y, Mao X, Zhou N. P-hydroxy benzaldehyde facilitates reprogramming of reactive astrocytes into neurons via endogenous transcriptional regulation. Int J Neurosci 2023; 133:1096-1108. [PMID: 35321633 DOI: 10.1080/00207454.2022.2049775] [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: 12/20/2021] [Accepted: 02/21/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cerebral ischemia leads to linguistic and motor dysfunction, as the death of neurons in ischemic core is permanent and non-renewable. An innovative avenue is to induce and/or facilitate reprogramming of adjacent astrocytes into neurons to replace the lost neurons and re-establish brain homeostasis. PURPOSE This study aimed to investigate whether the p-hydroxy benzaldehyde (p-HBA), a phenolic compound isolated from Gastrodia elata Blume, could facilitate the reprogramming of oxygen-glucose deprivation/reperfusion (OGD/R)-damaged astrocytes into neurons. STUDY DESIGN/METHODS The primary parenchymal astrocytes of rat were exposure to OGD and reperfusion with define culture medium. Cells were then incubated with different concentration of p-HBA (1, 10, 100, 400 μM) and collected at desired time point for reprogramming process analysis. RESULTS OGD/R could elicit endogenous neurogenic program in primary parenchymal astrocytes of rat under define culture condition, and these so-called reactive astrocytes could be reprogrammed into neurons. However, the neonatal neurons produced by this endogenous procedure could not develop into mature neurons, and the conversion rate was only 1.9%. Treatment of these reactive astrocytes with p-HBA could successfully promote the conversion rate to 6.1%, and the neonatal neurons could develop into mature neurons within 14 days. Further analysis showed that p-HBA down-regulated the Notch signal component genes Dll1, Hes1 and SOX2, while the transcription factor NeuroD1 was up-regulated. CONCLUSION The results of this study demonstrated that p-HBA facilitated the astrocyte-to-neuron conversion. This chemical reprogramming was mediated by inhibition of Notch1 signaling pathway and transcriptional activation of NeuroD1.
Collapse
Affiliation(s)
- Xin Li
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine
| | - Ruoxi Fan
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine
| | - Jianming Xiang
- Department of Neurosurgery, Medical School, University of Michigan, MI, USA
| | - Yajin Yuan
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine
| | - Xiaojian Mao
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine
| | - Ningna Zhou
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine
| |
Collapse
|
47
|
Hong L, Hong S, Zhang X. Expression and Functional Analysis of core stemness factors OSKM (OCT4, SOX2, KLF4, and MYC) in Pan-cancer. Medicine (Baltimore) 2023; 102:e36433. [PMID: 38050242 PMCID: PMC10695605 DOI: 10.1097/md.0000000000036433] [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: 09/11/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
The dedifferentiation process of tumorigenesis and somatic cell reprogramming has some commonness and differences, which is the key question to cancer therapeutic strategy and stem cell applications. To further explore the commonalities and variance between carcinogenesis and induced pluripotent stem cell reprogramming, we investigated the role of stemness factors OSKM (OCT4, SOX2, KLF4, and MYC) in the pan-cancer process using public clinical data. Expression of OSKM in human pan-cancer was analyzed via the Genotype Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) database based on the RNA-seq data of tissues. The correlation of expression between OSKM genes was analyzed via the Tumor Immune Evaluation Resource (TIMER) database, while the STRING tool was used to construct the protein-protein interaction network for OSKM. Prognostic impact of OSKM in pan-cancer was analyzed by Cox proportional hazards regression model. The relationships between OSKM and tumor stemness, tumor microenvironment and immune checkpoint and were performed by Sangerbox platform using Pearson correlation analysis. Our results showed that OSKM were universally expressed and significantly altered in tumors compared with adjacent normal tissues in most tumor types. In addition, correlation analysis revealed the relevance of OSKM genes to patient prognosis, cancer cell stemness, tumor microenvironment or immune checkpoint. However, there is little similarity between these genes in terms of how they function in each cancer type. This study elucidates the different roles of core stemness factors OSKM in pan-cancer, offering potential therapeutic targets for novel anti-cancer strategies and knowledge to minimize the potential carcinogenic effects during stem cell transplantation.
Collapse
Affiliation(s)
- Liwei Hong
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Medical College, Nanchang Institute of Technology, Nanchang, China
- Xiamen Clinical Research Center for Perinatal Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Sijie Hong
- Department of Ultrasound, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueqin Zhang
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
48
|
Ren K, Vickers R, Murillo J, Ruparel NB. Revolutionizing orofacial pain management: the promising potential of stem cell therapy. FRONTIERS IN PAIN RESEARCH 2023; 4:1239633. [PMID: 38028430 PMCID: PMC10679438 DOI: 10.3389/fpain.2023.1239633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Orofacial pain remains a significant health issue in the United States. Pain originating from the orofacial region can be composed of a complex array of unique target tissue that contributes to the varying success of pain management. Long-term use of analgesic drugs includes adverse effects such as physical dependence, gastrointestinal bleeding, and incomplete efficacy. The use of mesenchymal stem cells for their pain relieving properties has garnered increased attention. In addition to the preclinical and clinical results showing stem cell analgesia in non-orofacial pain, studies have also shown promising results for orofacial pain treatment. Here we discuss the outcomes of mesenchymal stem cell treatment for pain and compare the properties of stem cells from different tissues of origin. We also discuss the mechanism underlying these analgesic/anti-nociceptive properties, including the role of immune cells and the endogenous opioid system. Lastly, advancements in the methods and procedures to treat patients experiencing orofacial pain with mesenchymal stem cells are also discussed.
Collapse
Affiliation(s)
- Ke Ren
- Department of Pain and Neural Sciences, University of Maryland, Baltimore, MD, United States
| | - Russel Vickers
- Clinical Stem Cells Pty Ltd., Sydney, NSW, Australia
- Oral Health Center, School of Dentistry, Faculty of Health and Behavioural Sciences, The University of Queensland, Brisbane, QLD, Australia
- Institute for Glycomics, Griffith University Queensland, Southport, QLD, Australia
| | - Josue Murillo
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Nikita B. Ruparel
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
49
|
Mehra V, Chhetri JB, Ali S, Roddie C. The Emerging Role of Induced Pluripotent Stem Cells as Adoptive Cellular Immunotherapeutics. BIOLOGY 2023; 12:1419. [PMID: 37998018 PMCID: PMC10669440 DOI: 10.3390/biology12111419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/25/2023]
Abstract
Adoptive cell therapy (ACT) has transformed the treatment landscape for cancer and infectious disease through the investigational use of chimeric antigen receptor T-cells (CAR-Ts), tumour-infiltrating lymphocytes (TILs) and viral-specific T-cells (VSTs). Whilst these represent breakthrough treatments, there are subsets of patients who fail to respond to autologous ACT products. This is frequently due to impaired patient T-cell function or "fitness" as a consequence of prior treatments and age, and can be exacerbated by complex manufacturing protocols. Further, the manufacture of autologous, patient-specific products is time-consuming, expensive and non-standardised. Induced pluripotent stem cells (iPSCs) as an allogeneic alternative to patient-specific products can potentially overcome the issues outlined above. iPSC technology provides an unlimited source of rejuvenated iPSC-derived T-cells (T-iPSCs) or natural killer (NK) cells (NK-iPSCs), and in the context of the growing field of allogeneic ACT, iPSCs have enormous potential as a platform for generating off-the-shelf, standardised, "fit" therapeutics for patients. In this review, we evaluate current and future applications of iPSC technology in the CAR-T/NK, TIL and VST space. We discuss current and next-generation iPSC manufacturing protocols, and report on current iPSC-based adoptive therapy clinical trials to elucidate the potential of this technology as the future of ACT.
Collapse
Affiliation(s)
| | | | | | - Claire Roddie
- Research Department of Haematology, Cancer Institute, University College London, Paul O’Gorman Building, London WCIE 6DD, UK
| |
Collapse
|
50
|
Yu L, Wen H, Liu C, Wang C, Yu H, Zhang K, Han Q, Liu Y, Han Z, Li Z, Liu N. Embryonic stem cell-derived extracellular vesicles rejuvenate senescent cells and antagonize aging in mice. Bioact Mater 2023; 29:85-97. [PMID: 37449253 PMCID: PMC10336196 DOI: 10.1016/j.bioactmat.2023.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/11/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
Aging is a degenerative process that leads to tissue dysfunction and death. Embryonic stem cells (ESCs) have great therapeutic potential for age-related diseases due to their capacity for self-renewal and plasticity. However, the use of ESCs in clinical treatment is limited by immune rejection, tumourigenicity and ethical issues. ESC-derived extracellular vesicles (EVs) may provide therapeutic effects that are comparable to those of ESCs while avoiding unwanted effects. Here, we fully evaluate the role of ESC-EVs in rejuvenation in vitro and in vivo. Using RNA sequencing (RNA-Seq) and microRNA sequencing (miRNA-Seq) screening, we found that miR-15b-5p and miR-290a-5p were highly enriched in ESC-EVs, and induced rejuvenation by silencing the Ccn2-mediated AKT/mTOR pathway. These results demonstrate that miR-15b-5p and miR-290a-5p function as potent activators of rejuvenation mediated by ESC-EVs. The rejuvenating effect of ESC-EVs was further investigated in vivo by injection into aged mice. The results showed that ESC-EVs successfully ameliorated the pathological age-related phenotypes and rescued the transcriptome profile of aged mice. Our findings demonstrate that ESC-EVs treatment can rejuvenate senescence both in vitro and in vivo and suggest the therapeutic potential of ESC-EVs as a novel cell-free alternative to ESCs for age-related diseases.
Collapse
Affiliation(s)
- Lu Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Hang Wen
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Chen Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Huaxin Yu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Kaiyue Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qingsheng Han
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yue Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhongchao Han
- Institute of Stem Cells, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd, Tianjin, 301700, China
| | - Zongjin Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Na Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences Nankai University, Tianjin, 300071, China
| |
Collapse
|