1
|
de Silva L, van den Beucken JJJP, Rosenberg AJWP, Longoni A, Gawlitta D. Unraveling devitalization: its impact on immune response and ectopic bone remodeling from autologous and allogeneic callus mimics. Stem Cells Transl Med 2024; 13:1086-1100. [PMID: 39276211 PMCID: PMC11555479 DOI: 10.1093/stcltm/szae063] [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: 01/18/2024] [Accepted: 07/12/2024] [Indexed: 09/16/2024] Open
Abstract
Endochondral bone regeneration is a promising approach in regenerative medicine. Callus mimics (CMs) are engineered and remodeled into bone tissue upon implantation. The long-term objective is to fabricate a sustainable off-the-shelf treatment option for patients. Devitalization was introduced to facilitate storage and using allogeneic (donor) cells would further propel the off-the-shelf approach. However, allogeneic CMs for bone regeneration pose a potential antigenicity concern. Here, we explored the impact of devitalization on antigenicity and osteoinductive bone formation when implanting syngeneic or allogeneic CM in a vital or devitalized state. For this, we implanted chondrogenically differentiated rat-derived mesenchymal stromal cells using an allogeneic immunocompetent ectopic rat model. Vital syngeneic CMs demonstrated the highest bone formation, and vital allogeneic CMs showed the lowest bone formation, while both devitalized CMs showed comparable intermediate levels of bone formation. Preceding bone formation, the level of tartrate-resistant acid phosphatase staining at 7 and 14 days was proportional to the level of eventual bone formation. No differences were observed for local innate immune responses at any time point before or after bone formation. In contrast, allogeneic CMs elicit a mild adaptive immune response, which still permits bone formation in an immunocompetent environment, albeit at a reduced rate compared to the autologous living counterpart. Overall, devitalization delays bone formation when autologous CMs are implanted, whereas it accelerates bone formation in allogeneic CMs, highlighting the potential of this approach for achieving off-the-shelf treatment.
Collapse
Affiliation(s)
- Leanne de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht 3508 GA, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht 3584 CT, The Netherlands
| | | | - Antoine J W P Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht 3508 GA, The Netherlands
| | - Alessia Longoni
- Regenerative Medicine Center Utrecht, Utrecht 3584 CT, The Netherlands
- Department of Orthopedics, University Medical Center Utrecht, Utrecht University, Utrecht 3508 GA, The Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht 3508 GA, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht 3584 CT, The Netherlands
| |
Collapse
|
2
|
Li Y, Wang T, Li X, Li W, Lei Y, Shang Q, Zheng Z, Fang J, Cao L, Yu D, Meng Z, Zhang S, Liu R, Liu C, Xu C, Ding Y, Chen Y, Candi E, Melino G, Wang Y, Shi Y, Shao C. SOD2 promotes the immunosuppressive function of mesenchymal stem cells at the expense of adipocyte differentiation. Mol Ther 2024; 32:1144-1157. [PMID: 38310354 PMCID: PMC11163202 DOI: 10.1016/j.ymthe.2024.01.031] [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: 05/14/2023] [Revised: 11/28/2023] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
The potent immunomodulatory function of mesenchymal stem/stromal cells (MSCs) elicited by proinflammatory cytokines IFN-γ and TNF-α (IT) is critical to resolve inflammation and promote tissue repair. However, little is known about how the immunomodulatory capability of MSCs is related to their differentiation competency in the inflammatory microenvironment. In this study, we demonstrate that the adipocyte differentiation and immunomodulatory function of human adipose tissue-derived MSCs (MSC(AD)s) are mutually exclusive. Mitochondrial reactive oxygen species (mtROS), which promote adipocyte differentiation, were decreased in MSC(AD)s due to IT-induced upregulation of superoxide dismutase 2 (SOD2). Furthermore, knockdown of SOD2 led to enhanced adipogenic differentiation but reduced immunosuppression capability of MSC(AD)s. Interestingly, the adipogenic differentiation was associated with increased mitochondrial biogenesis and upregulation of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PPARGC1A/PGC-1α) expression. IT inhibited PGC-1α expression and decreased mitochondrial mass but promoted glycolysis in an SOD2-dependent manner. MSC(AD)s lacking SOD2 were compromised in their therapeutic efficacy in DSS-induced colitis in mice. Taken together, these findings indicate that the adipogenic differentiation and immunomodulation of MSC(AD)s may compete for resources in fulfilling the respective biosynthetic needs. Blocking of adipogenic differentiation by mitochondrial antioxidant may represent a novel strategy to enhance the immunosuppressive activity of MSCs in the inflammatory microenvironment.
Collapse
Affiliation(s)
- Yanan Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tingting Wang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Xiaolei Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Wen Li
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yan Lei
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Qianwen Shang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Zhiyuan Zheng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Jiankai Fang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Lijuan Cao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Daojiang Yu
- The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhenzhen Meng
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Shengchao Zhang
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Chunxiao Liu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Chenchang Xu
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yayun Ding
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Yongjing Chen
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China; Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Suzhou Medical College, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
3
|
Mahmoud M, Abdel-Rasheed M, Galal ER, El-Awady RR. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev Rep 2024; 20:175-205. [PMID: 37962697 PMCID: PMC10799834 DOI: 10.1007/s12015-023-10654-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Human adipose tissue-derived stem/stromal cells (hASCs) are adult multipotent mesenchymal stem/stromal cells with immunomodulatory capacities. Here, we present up-to-date knowledge on the impact of different experimental and donor-related factors on hASC immunoregulatory functions in vitro. The experimental determinants include the immunological status of hASCs relative to target immune cells, contact vs. contactless interaction, and oxygen tension. Factors such as the ratio of hASCs to immune cells, the cellular context, the immune cell activation status, and coculture duration are also discussed. Conditioning of hASCs with different approaches before interaction with immune cells, hASC culture in xenogenic or xenofree culture medium, hASC culture in two-dimension vs. three-dimension with biomaterials, and the hASC passage number are among the experimental parameters that greatly may impact the hASC immunosuppressive potential in vitro, thus, they are also considered. Moreover, the influence of donor-related characteristics such as age, sex, and health status on hASC immunomodulation in vitro is reviewed. By analysis of the literature studies, most of the indicated determinants have been investigated in broad non-standardized ranges, so the results are not univocal. Clear conclusions cannot be drawn for the fine-tuned scenarios of many important factors to set a standard hASC immunopotency assay. Such variability needs to be carefully considered in further standardized research. Importantly, field experts' opinions may help to make it clearer.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| | - Eman Reda Galal
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Rehab R El-Awady
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
4
|
Zhang L, Hoyos IA, Zubler C, Rieben R, Constantinescu M, Olariu R. Challenges and opportunities in vascularized composite allotransplantation of joints: a systematic literature review. Front Immunol 2023; 14:1179195. [PMID: 37275912 PMCID: PMC10235447 DOI: 10.3389/fimmu.2023.1179195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Background Joint allotransplantation (JA) within the field of vascularized composite allotransplantation (VCA) holds great potential for functional and non-prosthetic reconstruction of severely damaged joints. However, clinical use of JA remains limited due to the immune rejection associated with all forms of allotransplantation. In this study, we aim to provide a comprehensive overview of the current state of JA through a systematic review of clinical, animal, and immunological studies on this topic. Methods We conducted a systematic literature review in accordance with the PRISMA guidelines to identify relevant articles in PubMed, Cochrane Library, and Web of Science databases. The results were analyzed, and potential future prospects were discussed in detail. Results Our review included 14 articles describing relevant developments in JA. Currently, most JA-related research is being performed in small animal models, demonstrating graft survival and functional restoration with short-term immunosuppression. In human patients, only six knee allotransplantations have been performed to date, with all grafts ultimately failing and a maximum graft survival of 56 months. Conclusion Research on joint allotransplantation has been limited over the last 20 years due to the rarity of clinical applications, the complex nature of surgical procedures, and uncertain outcomes stemming from immune rejection. However, the key to overcoming these challenges lies in extending graft survival and minimizing immunosuppressive side effects. With the emergence of new immunosuppressive strategies, the feasibility and clinical potential of vascularized joint allotransplantation warrants further investigation.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Plastic and Reconstructive Surgery, Plastic and Reconstructive Surgery Center, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Isabel Arenas Hoyos
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Cédric Zubler
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Robert Rieben
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Mihai Constantinescu
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
| | - Radu Olariu
- Department of Plastic and Hand Surgery, Inselspital University Hospital Bern, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Sung TC, Maitiruze K, Pan J, Gong J, Bai Y, Pan X, Higuchi A. Universal and hypoimmunogenic pluripotent stem cells for clinical usage. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:271-296. [PMID: 37678974 DOI: 10.1016/bs.pmbts.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
It is urgent to prepare and store large numbers of clinical trial grade human pluripotent stem (hPS) cells for off-the-shelf use in stem cell therapies. However, stem cell banks, which store off-the-shelf stem cells, need financial support and large amounts of technicians for daily cell maintenance. Therefore, it is valuable to create "universal" or "hypoimmunogenic" hPS cells with genome editing engineering by knocking in or out immune-related genes. Only a small number of universal or hypoimmunogenic hPS cell lines should be needed to store for off-the-shelf usage and reduce the large amounts of instruments, consumables and technicians. In this article, we consider how to create hypoimmunogenic or universal hPS cells as well as the demerits of the technology. β2-Microglobulin-knockout hPS cells did not harbor human leukocyte antigen (HLA)-expressing class I cells but led to the activation of natural killer cells. To escape the activities of macrophages and natural killer cells, homozygous hPS cells having a single allele of an HLA class I gene, such as HLA-C, were proposed. Major HLA class Ia molecules were knocked out, and CD47, HLA-G and PD-L1 were knocked in hPS cells utilizing CRISPR/Cas9 genome editing. Finally, some researchers are trying to generate universal hPS cells without genome editing. The cells evaded the activation of not only T cells but also macrophages and natural killer cells. These universal hPS cells have high potential for application in cell therapy.
Collapse
Affiliation(s)
- Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Kailibinuer Maitiruze
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Jiandong Pan
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Jian Gong
- Department of Laboratory Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital, Wenzhou Medical University, The First Affiliated Hospital Area, Wenzhou, Zhejiang, P.R. China
| | - Xiaodong Pan
- Department of Urology, The First Affiliated Hospital, Wenzhou Medical University, The First Affiliated Hospital Area, Wenzhou, Zhejiang, P.R. China
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Chemical and Materials Engineering, National Central University, Jhongli, Taoyuan, Taiwan.
| |
Collapse
|
6
|
Garrity C, Arzi B, Haus B, Lee CA, Vapniarsky N. A Fresh Glimpse into Cartilage Immune Privilege. Cartilage 2022; 13:119-132. [PMID: 36250484 PMCID: PMC9924976 DOI: 10.1177/19476035221126349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The increasing prevalence of degenerative cartilage disorders in young patients is a growing public concern worldwide. Cartilage's poor innate regenerative capacity has inspired the exploration and development of cartilage replacement treatments such as tissue-engineered cartilages and osteochondral implants as potential solutions to cartilage loss. The clinical application of tissue-engineered implants is hindered by the lack of long-term follow-up demonstrating efficacy, biocompatibility, and bio-integration. The historically reported immunological privilege of cartilage tissue was based on histomorphological observations pointing out the lack of vascularity and the presence of a tight extracellular matrix. However, clinical studies in humans and animals do not unequivocally support the immune-privilege theory. More in-depth studies on cartilage immunology are needed to make clinical advances such as tissue engineering more applicable. This review analyzes the literature that supports and opposes the concept that cartilage is an immune-privileged tissue and provides insight into mechanisms conferring various degrees of immune privilege to other, more in-depth studied tissues such as testis, eyes, brain, and cancer.
Collapse
Affiliation(s)
- Carissa Garrity
- Department of Pathology, Microbiology
and Immunology, University of California, Davis, Davis, CA, USA
| | - Boaz Arzi
- Department of Surgical and Radiological
Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA,
USA
| | - Brian Haus
- Department of Orthopaedic Surgery,
University of California Davis Medical Center, Sacramento, CA, USA
| | - Cassandra A. Lee
- Department of Orthopaedic Surgery,
University of California Davis Medical Center, Sacramento, CA, USA
| | - Natalia Vapniarsky
- Department of Pathology, Microbiology
and Immunology, University of California, Davis, Davis, CA, USA,Natalia Vapniarsky, Department of
Pathology, Microbiology and Immunology, University of California, Davis, One
Shields Avenue, Davis, CA 95616-5270, USA.
| |
Collapse
|
7
|
Silver Nanoparticles Biocomposite Films with Antimicrobial Activity: In Vitro and In Vivo Tests. Int J Mol Sci 2022; 23:ijms231810671. [PMID: 36142584 PMCID: PMC9503464 DOI: 10.3390/ijms231810671] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Overuse of antimicrobials by the population has contributed to genetic modifications in bacteria and development of antimicrobial resistance, which is very difficult to combat nowadays. To solve this problem, it is necessary to develop new systems for the administration of antimicrobial active principles. Biocomposite systems containing silver nanoparticles can be a good medical alternative. In this context, the main objective of this study was to obtain a complex system in the form of a biocomposite film with antimicrobial properties based on chitosan, poly (vinyl alcohol) and silver nanoparticles. This new system was characterized from a structural and morphological point of view. The swelling degree, the mechanical properties and the efficiency of loading and release of an anti-inflammatory drug were also evaluated. The obtained biocomposite films are biocompatibles, this having been demonstrated by in vitro tests on HDFa cell lines, and have antimicrobial activity against S. aureus. The in vivo tests, carried out on rabbit subjects, highlighted the fact that signs of reduced fibrosis were specific to the C2P4.10.Ag1-IBF film sample, demonstrated by: intense expression of TNFAIP8 factors; as an anti-apoptotic marker, MHCII that favors immune cooperation among local cells; αSMA, which marks the presence of myofibroblasts involved in approaching the interepithelial spaces for epithelialization; and reduced expression of the Cox2 indicator of inflammation, Col I.
Collapse
|
8
|
Evaluation of a Novel Thiol–Norbornene-Functionalized Gelatin Hydrogel for Bioprinting of Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:ijms23147939. [PMID: 35887286 PMCID: PMC9321464 DOI: 10.3390/ijms23147939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/13/2022] [Accepted: 07/16/2022] [Indexed: 12/04/2022] Open
Abstract
Introduction: Three-dimensional bioprinting can be considered as an advancement of the classical tissue engineering concept. For bioprinting, cells have to be dispersed in hydrogels. Recently, a novel semi-synthetic thiolene hydrogel system based on norbornene-functionalized gelatin (GelNB) and thiolated gelatin (GelS) was described that resulted in the photoclick hydrogel GelNB/GelS. In this study, we evaluated the printability and biocompatibility of this hydrogel system towards adipose-tissue-derived mesenchymal stem cells (ASCs). Methods: GelNB/GelS was synthesized with three different crosslinking densities (low, medium and high), resulting in different mechanical properties with moduli of elasticity between 206 Pa and 1383 Pa. These hydrogels were tested for their biocompatibility towards ASCs in terms of their viability, proliferation and differentiation. The extrusion-based bioprinting of ASCs in GelNB/GelS-high was performed to manufacture three-dimensional cubic constructs. Results: All three hydrogels supported the viability, proliferation and chondrogenic differentiation of ASCs to a similar extent. The adipogenic differentiation of ASCs was better supported by the softer hydrogel (GelNB/GelS-low), whereas the osteogenic differentiation was more pronounced in the harder hydrogel (GelNB/GelS-high), indicating that the differentiation fate of ASCs can be influenced via the adaption of the mechanical properties of the GelNB/GelS system. After the ex vivo chondrogenic differentiation and subcutaneous implantation of the bioprinted construct into immunocompromised mice, the production of negatively charged sulfated proteoglycans could be observed with only minimal inflammatory signs in the implanted material. Conclusions: Our results indicate that the GelNB/GelS hydrogels are very well suited for the bioprinting of ASCs and may represent attractive hydrogels for subsequent in vivo tissue engineering applications.
Collapse
|
9
|
Tronik-Le Roux D, Daouya M, Jacquier A, Schenowitz C, Desgrandchamps F, Rouas-Freiss N, Carosella ED. The HLA-G immune checkpoint: a new immuno-stimulatory role for the α1-domain-deleted isoform. Cell Mol Life Sci 2022; 79:310. [PMID: 35596891 PMCID: PMC11072982 DOI: 10.1007/s00018-022-04359-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/25/2022] [Accepted: 05/09/2022] [Indexed: 11/03/2022]
Abstract
The heterogeneity of cancer cells, in part maintained via the expression of multiple isoforms, introduces significant challenges in designing effective therapeutic approaches. In this regard, isoforms of the immune checkpoint HLA-G have been found in most of the tumors analyzed, such as ccRCC, the most common human renal malignancy. In particular, HLA-G∆α1, which is the only HLA-G isoform described that lacks the α1 extracellular domain, has been newly identified in ccRCC and now here in trophoblasts. Using a cellular model expressing HLA-G∆α1, we have uncovered its specific and overlapping functional roles, relative to the main HLA-G isoform, i.e., the full-length HLA-G1. We found that HLA-G∆α1 has several particular features: (i) although possessing the α3 domain, it does not associate with β2-microglobulin; (ii) it may not present peptides to T cells due to absence of the peptide-binding groove; and (iii) it exerts immune-stimulatory activity towards peripheral blood NK and T cells, while all known isoforms of HLA-G are immune-inhibitory checkpoint molecules. Such immune-stimulatory properties of HLA-G∆α1 on the cytotoxic function of peripheral blood NK cells are individual dependent and are not exerted through the interaction with the known HLA-G receptor, ILT2. Importantly, we are faced here with a potential antitumor effect of an HLA-G isoform, opposed to the pro-tumor properties described for all other HLA-G isoforms, which should be taken into account in future therapeutic designs aimed at blocking this immune checkpoint.
Collapse
Affiliation(s)
- Diana Tronik-Le Roux
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France.
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France.
| | - Marina Daouya
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
| | - Alix Jacquier
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
| | - Chantal Schenowitz
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
| | - François Desgrandchamps
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
- Service d'Urologie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Nathalie Rouas-Freiss
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
| | - Edgardo D Carosella
- Atomic Energy and Alternative Energies Agency (CEA), Hematology and Immunology Research Division, Saint-Louis Hospital, 1, avenue Claude Vellefaux, 75010, Paris, France
- IRSL, UMRS 976, HIPI Unit, University of Paris, Paris, France
| |
Collapse
|
10
|
Wang J, Hao R, Jiang T, Guo X, Zhou F, Cao L, Gao F, Wang G, Wang J, Ning K, Zhong C, Chen X, Huang Y, Xu J, Gao S. Rebuilding hippocampus neural circuit with hADSC-derived neuron cells for treating ischemic stroke. Cell Biosci 2022; 12:40. [PMID: 35379347 PMCID: PMC8981707 DOI: 10.1186/s13578-022-00774-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 03/14/2022] [Indexed: 12/17/2022] Open
Abstract
Background Human adipose-derived stem cells (hADSCs) have been demonstrated to be a promising autologous stem cell source for treating various neuronal diseases. Our study indicated that hADSCs could be induced into neuron-like cells in a stepwise manner that are characterized by the positive expression of MAP2, SYNAPSIN 1/2, NF-200, and vGLUT and electrophysiological activity. We first primed hADSCs into neuron-like cells (hADSC-NCs) and then intracerebrally transplanted them into MCAO reperfusion mice to further explore their in vivo survival, migration, integration, fate commitment and involvement in neural circuit rebuilding. Results The hADSC-NCs survived well and transformed into MAP2-positive, Iba1- or GFAP-negative cells in vivo while maintaining some proliferative ability, indicated by positive Ki67 staining after 4 weeks. hADSC-NCs could migrate to multiple brain regions, including the cortex, hippocampus, striatum, and hypothalamus, and further differentiate into mature neurons, as confirmed by action potential elicitation and postsynaptic currents. With the aid of a cell suicide system, hADSC-NCs were proven to have functionally integrated into the hippocampal memory circuit, where they contributed to spatial learning and memory rescue, as indicated by LTP improvement and subsequent GCV-induced relapse. In addition to infarction size shrinkage and movement improvement, MCAO-reperfused mice showed bidirectional immune modulation, including inhibition of the local proinflammatory factors IL-1α, IL-1β, IL-2, MIP-1β and promotion proinflammatory IP-10, MCP-1, and enhancement of the anti-inflammatory factors IL-15. Conclusion Overall, hADSC-NCs used as an intermediate autologous cell source for treating stroke can rebuild hippocampus neuronal circuits through cell replacement. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00774-x.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Rui Hao
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai,, 200032,, China
| | - Tianfang Jiang
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Xuanxuan Guo
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Fei Zhou
- Department of Neurology, Third Affiliated Hospital of Navy Military Medical University, Shanghai, 200438, China
| | - Limei Cao
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China
| | - Fengjuan Gao
- Zhoupu Hospital, Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, 201318, China
| | - Guangming Wang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Juan Wang
- Department of Biotechnology and Molecular, Binzhou Medical College, Yantai, 264003, Shandong, People's Republic of China
| | - Ke Ning
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, 385A Glossop Road, Sheffield, S10 2HQ, UK
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xu Chen
- Department of Neurology, Shanghai Eighth People's Hospital Affiliated to Jiangsu University, Shanghai, 200233, China.
| | - Ying Huang
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Jun Xu
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Shane Gao
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
11
|
Expression of immunomodulatory and tissue regenerative biomarkers in human dental pulp derived-mesenchymal stem cells treated with curcumin: an in vitro study. Mol Biol Rep 2022; 49:4411-4420. [PMID: 35301656 DOI: 10.1007/s11033-022-07278-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/17/2021] [Accepted: 02/16/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Human Dental pulp derived-mesenchymal stem cells (hDP-MSCs) have the capability of selfrenewal, multipotency, as well as immunosuppressive properties. They are ideal candidates for regenerating damaged dental tissue and treating inflammation-related diseases. However, methods (such as genetic variation) to improve the immunomodulatory and regenerative efficiency of MSCs in different diseases still need to be developed. Curcumin (CUR) is known for its broad applications in regenerative medicine and the treatment of inflammatory disorders via its anti-inflammatory and anti-oxidant effects. This study was conducted to investigate the effect and underlying mechanisms of CUR on the immunomodulatory and regenerative function of hDP-MSCs and whether treating these cells with CUR can improve therapeutic efficacy. METHODS AND RESULTS hDP-MSCs were isolated from dental pulp and then treated with CUR. Cell viability rate was observed in hDP-MSCs after treatment of CUR by MTT assay. Real-time quantitative (RT-PCR) was applied to estimate the expression of immunomodulatory and regenerative genes after treatment of CUR. The RT-PCR results showed that VEGF-A and STAT3 markers were up-regulated while HLA-G5 and VCAM-1 markers were down-regulated by CUR (20 µM) treatment in hDP-MSCs (P < 0.001). Besides, this research indicated that there were no significant changes in the expressions of RelA and DSPP after 48 h (P = 0.33, P = 1). CONCLUSION Our findings demonstrate that CUR can enhance the immunomodulatory and regenerative effects of hDP-MSCs and improve their therapeutic efficacy. These findings can give an understanding of the mechanism for improving restorative and immunomodulatory activity in hDP-MSCs by curcumin.
Collapse
|
12
|
Uberti B, Plaza A, Henríquez C. Pre-conditioning Strategies for Mesenchymal Stromal/Stem Cells in Inflammatory Conditions of Livestock Species. Front Vet Sci 2022; 9:806069. [PMID: 35372550 PMCID: PMC8974404 DOI: 10.3389/fvets.2022.806069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) therapy has been a cornerstone of regenerative medicine in humans and animals since their identification in 1968. MSCs can interact and modulate the activity of practically all cellular components of the immune response, either through cell-cell contact or paracrine secretion of soluble mediators, which makes them an attractive alternative to conventional therapies for the treatment of chronic inflammatory and immune-mediated diseases. Many of the mechanisms described as necessary for MSCs to modulate the immune/inflammatory response appear to be dependent on the animal species and source. Although there is evidence demonstrating an in vitro immunomodulatory effect of MSCs, there are disparate results between the beneficial effect of MSCs in preclinical models and their actual use in clinical diseases. This discordance might be due to cells' limited survival or impaired function in the inflammatory environment after transplantation. This limited efficacy may be due to several factors, including the small amount of MSCs inoculated, MSC administration late in the course of the disease, low MSC survival rates in vivo, cryopreservation and thawing effects, and impaired MSC potency/biological activity. Multiple physical and chemical pre-conditioning strategies can enhance the survival rate and potency of MSCs; this paper focuses on hypoxic conditions, with inflammatory cytokines, or with different pattern recognition receptor ligands. These different pre-conditioning strategies can modify MSCs metabolism, gene expression, proliferation, and survivability after transplantation.
Collapse
Affiliation(s)
- Benjamin Uberti
- Instituto de Ciencias Clínicas, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anita Plaza
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henríquez
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
- *Correspondence: Claudio Henríquez
| |
Collapse
|
13
|
Mestrallet G, Carosella ED, Martin MT, Rouas-Freiss N, Fortunel NO, LeMaoult J. Immunosuppressive Properties of Epidermal Keratinocytes Differ According to Their Immaturity Status. Front Immunol 2022; 13:786859. [PMID: 35222373 PMCID: PMC8878806 DOI: 10.3389/fimmu.2022.786859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/14/2022] [Indexed: 12/04/2022] Open
Abstract
Preservation of a functional keratinocyte stem cell pool is essential to ensure the long-term maintenance of epidermis integrity, through continuous physiological renewal and regeneration in case of injury. Protecting stem cells from inflammation and immune reactions is thus a critical issue that needs to be explored. Here, we show that the immature CD49fhigh precursor cell fraction from interfollicular epidermis keratinocytes, comprising stem cells and progenitors, is able to inhibit CD4+ T-cell proliferation. Of note, both the stem cell-enriched CD49fhigh/EGFRlow subpopulation and the less immature CD49fhigh/EGFRhigh progenitors ensure this effect. Moreover, we show that HLA-G and PD-L1 immune checkpoints are overexpressed in CD49fhigh precursors, as compared to CD49flow differentiated keratinocytes. This potency may limit immune reactions against immature precursors including stem cells, and protect them from exacerbated inflammation. Further exploring this correlation between immuno-modulation and immaturity may open perspectives in allogenic cell therapies.
Collapse
Affiliation(s)
- Guillaume Mestrallet
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France
- Université Paris-Saclay, Saint-Aubin, France
| | - Edgardo D. Carosella
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, IRSL, Université Paris, Paris, France
| | - Michele T. Martin
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France
- Université Paris-Saclay, Saint-Aubin, France
| | - Nathalie Rouas-Freiss
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, IRSL, Université Paris, Paris, France
| | - Nicolas O. Fortunel
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France
- Université Paris-Saclay, Saint-Aubin, France
- *Correspondence: Joel LeMaoult, ; Nicolas O. Fortunel,
| | - Joel LeMaoult
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, Paris, France
- U976 HIPI Unit, IRSL, Université Paris, Paris, France
- *Correspondence: Joel LeMaoult, ; Nicolas O. Fortunel,
| |
Collapse
|
14
|
Fahy N, Palomares Cabeza V, Lolli A, Witte-Bouma J, Merino A, Ridwan Y, Wolvius EB, Hoogduijn MJ, Farrell E, Brama PAJ. Chondrogenically Primed Human Mesenchymal Stem Cells Persist and Undergo Early Stages of Endochondral Ossification in an Immunocompetent Xenogeneic Model. Front Immunol 2021; 12:715267. [PMID: 34659205 PMCID: PMC8515138 DOI: 10.3389/fimmu.2021.715267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Tissue engineering approaches using progenitor cells such as mesenchymal stromal cells (MSCs) represent a promising strategy to regenerate bone. Previous work has demonstrated the potential of chondrogenically primed human MSCs to recapitulate the process of endochondral ossification and form mature bone in vivo, using immunodeficient xenogeneic models. To further the translation of such MSC-based approaches, additional investigation is required to understand the impact of interactions between human MSC constructs and host immune cells upon the success of MSC-mediated bone formation. Although human MSCs are considered hypoimmunogenic, the potential of chondrogenically primed human MSCs to induce immunogenic responses in vivo, as well as the efficacy of MSC-mediated ectopic bone formation in the presence of fully competent immune system, requires further elucidation. Therefore, the aim of this study was to investigate the capacity of chondrogenically primed human MSC constructs to persist and undergo the process of endochondral ossification in an immune competent xenogeneic model. Chondrogenically differentiated human MSC pellets were subcutaneously implanted to wild-type BALB/c mice and retrieved at 2 and 12 weeks post-implantation. The percentages of CD4+ and CD8+ T cells, B cells, and classical/non-classical monocyte subsets were not altered in the peripheral blood of mice that received chondrogenic MSC constructs compared to sham-operated controls at 2 weeks post-surgery. However, MSC-implanted mice had significantly higher levels of serum total IgG compared to sham-operated mice at this timepoint. Flow cytometric analysis of retrieved MSC constructs identified the presence of T cells and macrophages at 2 and 12 weeks post-implantation, with low levels of immune cell infiltration to implanted MSC constructs detected by CD45 and CD3 immunohistochemical staining. Despite the presence of immune cells in the tissue, MSC constructs persisted in vivo and were not degraded/resorbed. Furthermore, constructs became mineralised, with longitudinal micro-computed tomography imaging revealing an increase in mineralised tissue volume from 4 weeks post-implantation until the experimental endpoint at 12 weeks. These findings indicate that chondrogenically differentiated human MSC pellets can persist and undergo early stages of endochondral ossification following subcutaneous implantation in an immunocompetent xenogeneic model. This scaffold-free model may be further extrapolated to provide mechanistic insight to osteoimmunological processes regulating bone regeneration and homeostasis.
Collapse
Affiliation(s)
- Niamh Fahy
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Orthopaedics and Sports Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Virginia Palomares Cabeza
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands.,Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Andrea Lolli
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Janneke Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Ana Merino
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Yanto Ridwan
- Department of Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Martin J Hoogduijn
- Transplantation Institute, Department of Internal Medicine, Erasmus Medical Center, Rotterdam, Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Center, Rotterdam, Netherlands
| | - Pieter A J Brama
- School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
15
|
Clumps of Mesenchymal Stem Cells/Extracellular Matrix Complexes Generated with Xeno-Free Chondro-Inductive Medium Induce Bone Regeneration via Endochondral Ossification. Biomedicines 2021; 9:biomedicines9101408. [PMID: 34680525 PMCID: PMC8533314 DOI: 10.3390/biomedicines9101408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/04/2021] [Accepted: 09/28/2021] [Indexed: 01/14/2023] Open
Abstract
Three-dimensional clumps of mesenchymal stem cells (MSCs)/extracellular matrix (ECM) complexes (C-MSCs) can be transplanted into tissue defect site with no artificial scaffold. Importantly, most bone formation in the developing process or fracture healing proceeds via endochondral ossification. Accordingly, this present study investigated whether C-MSCs generated with chondro-inductive medium (CIM) can induce successful bone regeneration and assessed its healing process. Human bone marrow-derived MSCs were cultured with xeno-free/serum-free (XF) growth medium. To obtain C-MSCs, confluent cells that had formed on the cellular sheet were scratched using a micropipette tip and then torn off. The sheet was rolled to make a round clump of cells. The cell clumps, i.e., C-MSCs, were maintained in XF-CIM. C-MSCs generated with XF-CIM showed enlarged round cells, cartilage matrix, and hypertrophic chondrocytes genes elevation in vitro. Transplantation of C-MSCs generated with XF-CIM induced successful bone regeneration in the SCID mouse calvaria defect model. Immunofluorescence staining for human-specific vimentin demonstrated that donor human and host mouse cells cooperatively contributed the bone formation. Besides, the replacement of the cartilage matrix into bone was observed in the early period. These findings suggested that cartilaginous C-MSCs generated with XF-CIM can induce bone regeneration via endochondral ossification.
Collapse
|
16
|
Krampera M, Le Blanc K. Mesenchymal stromal cells: Putative microenvironmental modulators become cell therapy. Cell Stem Cell 2021; 28:1708-1725. [PMID: 34624232 DOI: 10.1016/j.stem.2021.09.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
An exceptional safety profile has been shown in a large number of cell therapy clinical trials that use mesenchymal stromal cells (MSCs). However, reliable potency assays are still lacking to predict MSC immunosuppressive efficacy in the clinical setting. Nevertheless, MSCs are approved in Japan and Europe for the treatment of graft-versus-host and Crohn's fistular diseases, but not in the United States for any clinical indication. We discuss potential mechanisms of action for the therapeutic effects of MSC transplantation, experimental models that dissect tissue modulating function of MSCs, and approaches for identifying MSC effects in vivo by integrating biomarkers of disease and MSC activity.
Collapse
Affiliation(s)
- Mauro Krampera
- Section of Hematology and Bone Marrow Transplant Unit, Department of Medicine, University of Verona, Verona, Italy.
| | - Katarina Le Blanc
- Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet, Stockholm, Sweden; Center of Allogeneic Stem Cell Transplantation and Cellular Therapy (CAST), Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| |
Collapse
|
17
|
Mardomi A, Limoni SK, Rahbarghazi R, Mohammadi N, Khorashadizadeh M, Ranjbaran H, Nataj HH, Jafari N, Hasani B, Abediankenari S. PD-L1 overexpression conveys tolerance of mesenchymal stem cell-derived cardiomyocyte-like cells in an allogeneic mouse model. J Cell Physiol 2021; 236:6328-6343. [PMID: 33507552 DOI: 10.1002/jcp.30299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/28/2020] [Accepted: 01/13/2021] [Indexed: 01/14/2023]
Abstract
Although the autologously transplanted cells are immunologically durable, allogeneic cell transplantation is inevitable in a series of cases. Mesenchymal stem cells (MSCs) are one of the suitable candidates for cardiac tissue regeneration that have been shown to acquire immunogenicity concurrent with cardiomyogenic differentiation. The present study aimed to exploit PD-L1, as a key immunomodulatory checkpoint ligand to protect the MSCs-derived cardiomyocyte-like cells (CLCs) against the detrimental alloimmunity. Mouse bone marrow-derived MSCs were stably transduced to overexpress PD-L1. MSCs were in vitro differentiated into CLCs and the expressions of immunologic molecules were compared between MSCs and CLCs. The in vitro and in vivo allogeneic immune responses were also examined. The differentiated CLCs had higher expressions of MHC-I and CD80. Upon in vitro coculture with allogeneic splenocytes, CLCs caused more CD4+ and CD8+ T cell activation, lymphocyte proliferation, and interferon-γ (IFN-γ) release in comparison to MSCs. PD-L1 overexpression on CLCs decreased the activation of CD8+ T cells, proliferation of lymphocytes, and release of IFN-γ. The PD-L1-overexpressing CLCs elicited lower in vivo CD4+ and CD8+ T cell activation and reduced the anti-donor antibody response accompanied by increased durability and reduced T cell infiltration. The present study verified the potential of PD-L1 overexpression as a preparative strategy for the protection of allogeneic MSCs-derived CLCs against the detrimental alloreaction.
Collapse
Affiliation(s)
- Alireza Mardomi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shabanali K Limoni
- Department of Medical Biotechnology, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nabiallah Mohammadi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohsen Khorashadizadeh
- Department of Medical Biotechnology, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Ranjbaran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hadi H Nataj
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narjes Jafari
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Bahareh Hasani
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
18
|
Mestrallet G, Auvré F, Schenowitz C, Carosella ED, LeMaoult J, Martin MT, Rouas-Freiss N, Fortunel NO. Human Keratinocytes Inhibit CD4 + T-Cell Proliferation through TGFB1 Secretion and Surface Expression of HLA-G1 and PD-L1 Immune Checkpoints. Cells 2021; 10:cells10061438. [PMID: 34201301 PMCID: PMC8227977 DOI: 10.3390/cells10061438] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023] Open
Abstract
Human skin protects the body against infection and injury. This protection involves immune and epithelial cells, but their interactions remain largely unknown. Here, we show that cultured epidermal keratinocytes inhibit allogenic CD4+ T-cell proliferation under both normal and inflammatory conditions. Inhibition occurs through the secretion of soluble factors, including TGFB1 and the cell-surface expression of HLA-G1 and PD-L1 immune checkpoints. For the first time, we here describe the expression of the HLA-G1 protein in healthy human skin and its role in keratinocyte-driven tissue immunomodulation. The overexpression of HLA-G1 with an inducible vector increased the immunosuppressive properties of keratinocytes, opening up perspectives for their use in allogeneic settings for cell therapy.
Collapse
Affiliation(s)
- Guillaume Mestrallet
- CEA, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Francois Jacob Institute of Biology, DRF, 91000 Evry, France; (G.M.); (F.A.)
- Université Paris-Saclay, 91190 Saint-Aubin, France
| | - Frédéric Auvré
- CEA, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Francois Jacob Institute of Biology, DRF, 91000 Evry, France; (G.M.); (F.A.)
- Université Paris-Saclay, 91190 Saint-Aubin, France
| | - Chantal Schenowitz
- CEA, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, 75010 Paris, France; (C.S.); (E.D.C.)
- U976 HIPI Unit, IRSL, Université Paris, 75010 Paris, France
| | - Edgardo D. Carosella
- CEA, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, 75010 Paris, France; (C.S.); (E.D.C.)
- U976 HIPI Unit, IRSL, Université Paris, 75010 Paris, France
| | - Joel LeMaoult
- CEA, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, 75010 Paris, France; (C.S.); (E.D.C.)
- U976 HIPI Unit, IRSL, Université Paris, 75010 Paris, France
- Correspondence: (J.L.); (M.T.M.); (N.R.-F.); (N.O.F.); Tel.: +33-1-60-87-34-91 (M.T.M.); +33-1-57-27-68-01 (N.R.-F.); +33-1-60-87-34-92 (N.O.F.)
| | - Michèle T. Martin
- CEA, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Francois Jacob Institute of Biology, DRF, 91000 Evry, France; (G.M.); (F.A.)
- Université Paris-Saclay, 91190 Saint-Aubin, France
- Correspondence: (J.L.); (M.T.M.); (N.R.-F.); (N.O.F.); Tel.: +33-1-60-87-34-91 (M.T.M.); +33-1-57-27-68-01 (N.R.-F.); +33-1-60-87-34-92 (N.O.F.)
| | - Nathalie Rouas-Freiss
- CEA, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, 75010 Paris, France; (C.S.); (E.D.C.)
- U976 HIPI Unit, IRSL, Université Paris, 75010 Paris, France
- Correspondence: (J.L.); (M.T.M.); (N.R.-F.); (N.O.F.); Tel.: +33-1-60-87-34-91 (M.T.M.); +33-1-57-27-68-01 (N.R.-F.); +33-1-60-87-34-92 (N.O.F.)
| | - Nicolas O. Fortunel
- CEA, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Francois Jacob Institute of Biology, DRF, 91000 Evry, France; (G.M.); (F.A.)
- Université Paris-Saclay, 91190 Saint-Aubin, France
- Correspondence: (J.L.); (M.T.M.); (N.R.-F.); (N.O.F.); Tel.: +33-1-60-87-34-91 (M.T.M.); +33-1-57-27-68-01 (N.R.-F.); +33-1-60-87-34-92 (N.O.F.)
| |
Collapse
|
19
|
Gibler P, Gimble J, Hamel K, Rogers E, Henderson M, Wu X, Olesky S, Frazier T. Human Adipose-Derived Stromal/Stem Cell Culture and Analysis Methods for Adipose Tissue Modeling In Vitro: A Systematic Review. Cells 2021; 10:1378. [PMID: 34204869 PMCID: PMC8227575 DOI: 10.3390/cells10061378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Human adipose-derived stromal/stem cells (hASC) are widely used for in vitro modeling of physiologically relevant human adipose tissue. These models are useful for the development of tissue constructs for soft tissue regeneration and 3-dimensional (3D) microphysiological systems (MPS) for drug discovery. In this systematic review, we report on the current state of hASC culture and assessment methods for adipose tissue engineering using 3D MPS. Our search efforts resulted in the identification of 184 independent records, of which 27 were determined to be most relevant to the goals of the present review. Our results demonstrate a lack of consensus on methods for hASC culture and assessment for the production of physiologically relevant in vitro models of human adipose tissue. Few studies have assessed the impact of different 3D culture conditions on hASC adipogenesis. Additionally, there has been a limited use of assays for characterizing the functionality of adipose tissue in vitro. Results from this study suggest the need for more standardized culture methods and further analysis on in vitro tissue functionality. These will be necessary to validate the utility of 3D MPS as an in vitro model to reduce, refine, and replace in vivo experiments in the drug discovery regulatory process.
Collapse
Affiliation(s)
- Peyton Gibler
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Jeffrey Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Katie Hamel
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Emma Rogers
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Michael Henderson
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Spencer Olesky
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
20
|
Pethe P, Kale V. Placenta: A gold mine for translational research and regenerative medicine. Reprod Biol 2021; 21:100508. [PMID: 33930790 DOI: 10.1016/j.repbio.2021.100508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023]
Abstract
Stem cell therapy has gained much impetus in regenerative medicine due to some of the encouraging results obtained in the laboratory as well as in translational/clinical studies. Although stem cells are of various types and their therapeutic potential has been documented in several studies, mesenchymal stromal/stem cells (MSCs) have an edge, as in addition to being multipotent, these cells are easy to obtain and expand, pose fewer ethical issues, and possess immense regenerative potential when used in a scientifically correct manner. Currently, MSCs are being sourced from various tissues such as bone marrow, cord, cord blood, adipose tissue, dental tissue, etc., and, quite often, the choice depends on the availability of the source. One such rich source of tissue suitable for obtaining good quality MSCs in large numbers is the placenta obtained in a full-term delivery leading to a healthy child's birth. Several studies have demonstrated the regenerative potential of human placenta-derived MSCs (hPMSC), and most show that these MSCs possess comparable, in some instances, even better, therapeutic potential as that shown by human bone marrow-derived (hBMSC) or human umbilical cord-derived (hUC-MSC) MSCs. The placenta can be easily sourced from the OB/GYN department of any hospital, and if its derivatives such as hPMSC or their EVs are produced under GMP conditions, it could serve as a gold mine for translational/clinical research. Here, we have reviewed recent studies revealing the therapeutic potential of hPMSC and their extracellular vesicles (EVs) published over the past three years.
Collapse
Affiliation(s)
- Prasad Pethe
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, 412115, India.
| |
Collapse
|
21
|
Hansen M, Stahl L, Heider A, Hilger N, Sack U, Kirschner A, Cross M, Fricke S. Reduction of Graft-versus-Host-Disease in NOD.Cg-Prkdc scid Il2rg tm1Wjl/SzJ (NSG) Mice by Cotransplantation of Syngeneic Human Umbilical Cord-Derived Mesenchymal Stromal Cells. Transplant Cell Ther 2021; 27:658.e1-658.e10. [PMID: 33964513 DOI: 10.1016/j.jtct.2021.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 04/25/2021] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is one of the major complications following hematopoietic stem cell transplantation, which remains the sole curative therapy for many malignant diseases of the hematopoietic system. The immunomodulatory potential of mesenchymal stromal cells (MSCs) to treat GVHD is currently being tested in various preclinical and clinical trials. Because the results of the preclinical and clinical trials on the use of MSCs to treat GVHD have not been consistent, we analyzed the potential beneficial effects of syngeneic versus allogenic treatment, culture expansion of MSCs, and various MSC cell doses and time points of MSC transplantation in a murine GVHD model. We established the murine GVHD model based on the transplantation of umbilical cord blood-derived hematopoietic stem cells (UC-HSCs) and used this model to assess the therapeutic potential of umbilical cord blood-derived MSCs (UC-MSCs). The use of HSC and MSC populations derived from the same donor allowed us to exclude third-party cells and test the UC-HSCs and UC-MSCs in a matched setting. Moreover, we were able to compare various doses, transplantation time points, and the influence of culture expansion of MSCs on the impact of treatment. This resulted in 16 different treatment groups. The most efficient setting for treatment of UC-HSC-induced GVHD reactions was based on the simultaneous administration of 1 × 106 culture-expanded, syngeneically matched UC-MSCs. This therapy effectively reduced the number of CD8+ T cells in the blood, protected the mice from weight loss, and prolonged their survival until the end of observation period. Taken together, our data show beneficial effects of (1) syngeneic over allogeneic UC-HSCs and UC-MSCs, (2) culture-expanded cells over freshly isolated primary cells, (3) simultaneous over sequential administration, and (4) high doses of UC-MSCs. The animal model of GVHD established here is now available for more detailed studies, including a comparative analysis of the efficacy of MSCs derived from alternative sources, such as adipose tissue and bone marrow.
Collapse
Affiliation(s)
- Max Hansen
- Vita 34 AG, Leipzig, Germany; Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany.
| | - Lilly Stahl
- Tcell Tolerance GmbH, Leipzig, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | | | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Ulrich Sack
- Department of Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Andreas Kirschner
- Vita 34 AG, Leipzig, Germany; Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Michael Cross
- Department of Hematology and Cell Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
22
|
Rocha JLM, de Oliveira WCF, Noronha NC, Dos Santos NCD, Covas DT, Picanço-Castro V, Swiech K, Malmegrim KCR. Mesenchymal Stromal Cells in Viral Infections: Implications for COVID-19. Stem Cell Rev Rep 2021; 17:71-93. [PMID: 32895900 PMCID: PMC7476649 DOI: 10.1007/s12015-020-10032-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) constitute a heterogeneous population of stromal cells with immunomodulatory and regenerative properties that support their therapeutic use. MSCs isolated from many tissue sources replicate vigorously in vitro and maintain their main biological properties allowing their widespread clinical application. To date, most MSC-based preclinical and clinical trials targeted immune-mediated and inflammatory diseases. Nevertheless, MSCs have antiviral properties and have been used in the treatment of various viral infections in the last years. Here, we revised in detail the biological properties of MSCs and their preclinical and clinical applications in viral diseases, including the disease caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) infection (COVID-19). Notably, rapidly increasing numbers of MSC-based therapies for COVID-19 have recently been reported. MSCs are theoretically capable of reducing inflammation and promote lung regeneration in severe COVID-19 patients. We critically discuss the rationale, advantages and disadvantages of MSC-based therapies for viral infections and also specifically for COVID-19 and point out some directions in this field. Finally, we argue that MSC-based therapy may be a promising therapeutic strategy for severe COVID-19 and other emergent respiratory tract viral infections, beyond the viral infection diseases in which MSCs have already been clinically applied. Graphical Abstract ![]()
Collapse
Affiliation(s)
- José Lucas Martins Rocha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Waldir César Ferreira de Oliveira
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Nádia Cássia Noronha
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Natalia Cristine Dias Dos Santos
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Bioscience and Biotecnology Program, Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dimas Tadeu Covas
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Virgínia Picanço-Castro
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kamilla Swiech
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, Ribeirão Preto, 14040-903, São Paulo, Brazil
| | - Kelen Cristina Ribeiro Malmegrim
- Center for Cell-based Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil. .,School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café s/n, Ribeirão Preto, 14040-903, São Paulo, Brazil.
| |
Collapse
|
23
|
Wong KU, Zhang A, Akhavan B, Bilek MM, Yeo GC. Biomimetic Culture Strategies for the Clinical Expansion of Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2021. [PMID: 33599471 DOI: 10.1021/acsbiomaterials.0c01538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) typically require significant ex vivo expansion to achieve the high cell numbers required for research and clinical applications. However, conventional MSC culture on planar (2D) plastic surfaces has been shown to induce MSC senescence and decrease cell functionality over long-term proliferation, and usually, it has a high labor requirement, a high usage of reagents, and therefore, a high cost. In this Review, we describe current MSC-based therapeutic strategies and outline the important factors that need to be considered when developing next-generation cell expansion platforms. To retain the functional value of expanded MSCs, ex vivo culture systems should ideally recapitulate the components of the native stem cell microenvironment, which include soluble cues, resident cells, and the extracellular matrix substrate. We review the interplay between these stem cell niche components and their biological roles in governing MSC phenotype and functionality. We discuss current biomimetic strategies of incorporating biochemical and biophysical cues in MSC culture platforms to grow clinically relevant cell numbers while preserving cell potency and stemness. This Review summarizes the current state of MSC expansion technologies and the challenges that still need to be overcome for MSC clinical applications to be feasible and sustainable.
Collapse
Affiliation(s)
- Kuan Un Wong
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anyu Zhang
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Behnam Akhavan
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Marcela M Bilek
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
24
|
Bazzoni R, Takam Kamga P, Tanasi I, Krampera M. Extracellular Vesicle-Dependent Communication Between Mesenchymal Stromal Cells and Immune Effector Cells. Front Cell Dev Biol 2020; 8:596079. [PMID: 33240892 PMCID: PMC7677193 DOI: 10.3389/fcell.2020.596079] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are multipotent cells residing in the stromal tissues of the body and capable of promoting tissue repair and attenuating inflammatory processes through their immunomodulatory properties. Preclinical and clinical observations revealed that not only direct intercellular communication mediates MSC properties; in fact, a pivotal role is also played by the release of soluble and bioactive factors, such as cytokines, growth factor and extracellular vesicles (EVs). EVs are membrane-coated vesicles containing a large variety of bioactive molecules, including lipids, proteins, and nucleic acids, such as RNA. EVs release their contents into target cells, thus influencing cell fate through the control of intracellular processes. In addition, MSC-derived EVs can mediate modulatory effects toward different effector cells belonging to both innate and adaptive immunity. In this review, we will discuss the literature data concerning MSC-derived EVs, including the current standardized methods for their isolation and characterization, the mechanisms supporting their immunoregulatory properties, and their potential clinical application as alternative to MSC-based therapy for inflammatory reactions, such as graft-versus-host disease (GvHD).
Collapse
Affiliation(s)
- Riccardo Bazzoni
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Paul Takam Kamga
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
- EA4340-BCOH, Biomarker in Cancerology and Onco-Haematology, UVSQ, Université Paris Saclay, Boulogne-Billancourt, France
| | - Ilaria Tanasi
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Krampera
- Stem Cell Research Laboratory, Section of Hematology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
25
|
Longoni A, Pennings I, Cuenca Lopera M, van Rijen MHP, Peperzak V, Rosenberg AJWP, Levato R, Gawlitta D. Endochondral Bone Regeneration by Non-autologous Mesenchymal Stem Cells. Front Bioeng Biotechnol 2020; 8:651. [PMID: 32733861 PMCID: PMC7363768 DOI: 10.3389/fbioe.2020.00651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/27/2020] [Indexed: 12/31/2022] Open
Abstract
Mimicking endochondral bone formation is a promising strategy for bone regeneration. To become a successful therapy, the cell source is a crucial translational aspect. Typically, autologous cells are used. The use of non-autologous mesenchymal stromal cells (MSCs) represents an interesting alternative. Nevertheless, non-autologous, differentiated MSCs may trigger an undesired immune response, hampering bone regeneration. The aim of this study was to unravel the influence of the immune response on endochondral bone regeneration, when using xenogeneic (human) or allogeneic (Dark Agouti) MSCs. To this end, chondrogenically differentiated MSCs embedded in a collagen carrier were implanted in critical size femoral defects of immunocompetent Brown Norway rats. Control groups were included with syngeneic/autologous (Brown Norway) MSCs or a cell-free carrier. The amount of neo-bone formation was proportional to the degree of host-donor relatedness, as no full bridging of the defect was observed in the xenogeneic group whereas 2/8 and 7/7 bridges occurred in the allogeneic and the syngeneic group, respectively. One week post-implantation, the xenogeneic grafts were invaded by pro-inflammatory macrophages, T lymphocytes, which persisted after 12 weeks, and anti-human antibodies were developed. The immune response toward the allogeneic graft was comparable to the one evoked by the syngeneic implants, aside from an increased production of alloantibodies, which might be responsible for the more heterogeneous bone formation. Our results demonstrate for the first time the feasibility of using non-autologous MSC-derived chondrocytes to elicit endochondral bone regeneration in vivo. Nevertheless, the pronounced immune response and the limited bone formation observed in the xenogeneic group undermine the clinical relevance of this group. On the contrary, although further research on how to achieve robust bone formation with allogeneic cells is needed, they may represent an alternative to autologous transplantation.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - I Pennings
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| | - Marta Cuenca Lopera
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - M H P van Rijen
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Victor Peperzak
- Laboratory for Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - A J W P Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, Netherlands.,Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Regenerative Medicine Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
26
|
Voisin C, Cauchois G, Reppel L, Laroye C, Louarn L, Schenowitz C, Sonon P, Poras I, Wang V, D. Carosella E, Benkirane-Jessel N, Moreau P, Rouas-Freiss N, Bensoussan D, Huselstein C. Are the Immune Properties of Mesenchymal Stem Cells from Wharton's Jelly Maintained during Chondrogenic Differentiation? J Clin Med 2020; 9:jcm9020423. [PMID: 32033151 PMCID: PMC7073626 DOI: 10.3390/jcm9020423] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 01/13/2023] Open
Abstract
Background: Umbilical mesenchymal stem/stromal cells (MSCs), and especially those derived from Wharton’s jelly (WJ), are a promising engineering tool for tissue repair in an allogeneic context. This is due to their differentiation capacity and immunological properties, like their immunomodulatory potential and paracrine activity. Hence, these cells may be considered an Advanced Therapy Medicinal Product (ATMP). The purpose of this work was to differentiate MSCs from WJ (WJ-MSCs) into chondrocytes using a scaffold and to evaluate, in vitro, the immunomodulatory capacities of WJ-MSCs in an allogeneic and inflammatory context, mimicked by IFN-γ and TNF-α priming during the chondrogenic differentiation. Methods: Scaffolds were made from hydrogel composed by alginate enriched in hyaluronic acid (Alg/HA). Chondrogenic differentiation, immunological function, phenotype expression, but also secreted soluble factors were the different parameters followed during 28 days of culture. Results: During chondrocyte differentiation, even in an allogeneic context, WJ-MSCs remained unable to establish the immunological synapse or to induce T cell alloproliferation. Moreover, interestingly, paracrine activity and functional immunomodulation were maintained during cell differentiation. Conclusion: These results show that WJ-MSCs remained hypoimmunogenic and retained immunomodulatory properties even when they had undergone chondrocyte differentiation.
Collapse
Affiliation(s)
- Charlotte Voisin
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
- Correspondence: ; Tel.: +33-372-74-6585
| | - Ghislaine Cauchois
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
| | - Loïc Reppel
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire Banque de Tissus, 54500 Vandœuvre-lès-Nancy, France
| | - Caroline Laroye
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
- CHRU de Nancy, Unité de Thérapie Cellulaire Banque de Tissus, 54500 Vandœuvre-lès-Nancy, France
| | - Laetitia Louarn
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Chantal Schenowitz
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Paulin Sonon
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Isabelle Poras
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Valentine Wang
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
| | - Edgardo D. Carosella
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Nadia Benkirane-Jessel
- INSERM-UNISTRA UMR1260, Regenerative Nanomedicine laboratory, Faculté de Médecine, FMTS, Strasbourg CEDEX F-67085, France;
| | - Philippe Moreau
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Nathalie Rouas-Freiss
- CEA, DRF-Institut François Jacob, Service de Recherches en Hémato-Immunologie, Hopital Saint-Louis, 75010 Paris, France; (L.L.); (C.S.); (P.S.); (I.P.); (E.D.C.); (P.M.); (N.R.-F.)
- Université de Paris, CEA, U976 HIPI Unit (Human Immunology, Physiopathology, Immunotherapy), Institut de Recherche Saint-Louis, 75010 Paris, France
| | - Danièle Bensoussan
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- CHRU de Nancy, Unité de Thérapie Cellulaire Banque de Tissus, 54500 Vandœuvre-lès-Nancy, France
| | - Céline Huselstein
- UMR 7365 CNRS-Université de Lorraine, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Biopôle de l’Université de Lorraine, Campus brabois-santé, Faculté de Médecine, 9 Avenue de la Forêt de Haye, BP 184, 54500 Vandoeuvre-lès-nancy, France; (G.C.); (L.R.); (C.L.); (V.W.); (D.B.); (C.H.)
- UMS2008 IBSLor, Campus brabois-santé, 9 Avenue de la Forêt de Haye, BP20199, 54500 Vandoeuvre-lès-nancy, France
| |
Collapse
|
27
|
Jiang W, Xu J. Immune modulation by mesenchymal stem cells. Cell Prolif 2019; 53:e12712. [PMID: 31730279 PMCID: PMC6985662 DOI: 10.1111/cpr.12712] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/11/2019] [Accepted: 10/08/2019] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be derived from various adult tissues with multipotent and self‐renewal abilities. The characteristics of presenting no major ethical concerns, having low immunogenicity and possessing immune modulation functions make MSCs promising candidates for stem cell therapies. MSCs could promote inflammation when the immune system is underactivated and restrain inflammation when the immune system is overactivated to avoid self‐overattack. These cells express many immune suppressors to switch them from a pro‐inflammatory phenotype to an anti‐inflammatory phenotype, resulting in immune effector cell suppression and immune suppressor cell activation. We would discuss the mechanisms governing the immune modulation function of these cells in this review, especially the immune‐suppressive effects of MSCs.
Collapse
Affiliation(s)
- Wei Jiang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Anatomy, Histology & Developmental Biology, Health Science Center, Shenzhen University, Shenzhen, China.,Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
28
|
Xu T, Yu X, Yang Q, Liu X, Fang J, Dai X. Autologous Micro-Fragmented Adipose Tissue as Stem Cell-Based Natural Scaffold for Cartilage Defect Repair. Cell Transplant 2019; 28:1709-1720. [PMID: 31565996 PMCID: PMC6923561 DOI: 10.1177/0963689719880527] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Osteoarthritis (OA) poses a tough challenge worldwide. Adipose-derived stem cells (ASCs)
have been proved to play a promising role in cartilage repair. However, enzymatic
digestion, ex vivo culture and expansion, with significant senescence and decline in
multipotency, limit their application. The present study was designed to obtain
micro-fragmented adipose tissue (MFAT) through gentle mechanical force and determine the
effect of this stem cell-based natural scaffold on repair of full-thickness cartilage
defects. In this study, ASCs sprouted from MFAT were characterized by
multi-differentiation induction and flow cytometry. Scratch and transwell migration assays
were operated to determine whether MFAT could promote migration of chondrocytes in vitro.
In a rat model, cartilage defects were created on the femoral groove and treated with
intra-articular injection of MFAT or PBS for 6 weeks and 12 weeks (n =
12). At the time points, the degree of cartilage repair was evaluated by histological
staining, immunohistochemistry and scoring, respectively. Two unoperated age-matched
animals served as native controls. ASCs derived from MFAT possessed properties to
differentiate into adipocytes, osteocytes and chondrocytes, with expression of mesenchymal
stem cell markers (CD29, 44, 90) and no expression of hematopoietic markers (CD31, 34,
45). In addition, MFAT could significantly promote migration of chondrocytes. MFAT-treated
defects showed improved macroscopic appearance and histological evaluation compared with
PBS-treated defects at both time points. After 12 weeks of treatment, MFAT-treated defects
displayed regular surface, high amount of hyaline cartilage, intact subchondral bone
reconstruction and corresponding formation of type I, II, and VI collagen, which resembled
the normal cartilage. This study demonstrates the efficacy of MFAT on cartilage repair in
an animal model for the first time, and the utility of MFAT as a ready-to-use therapeutic
alternative to traditional stem cell therapy.
Collapse
Affiliation(s)
- Tengjing Xu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinning Yu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| | - Quanming Yang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaonan Liu
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghua Fang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| | - Xuesong Dai
- Department of Orthopaedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Department of Orthopaedic Surgery, Hangzhou Mingzhou Hospital (International Medical Center, Second Affiliated Hospital, Zhejiang University School of Medicine), Hangzhou, China
| |
Collapse
|
29
|
Characterization of Different Sources of Human MSCs Expanded in Serum-Free Conditions with Quantification of Chondrogenic Induction in 3D. Stem Cells Int 2019; 2019:2186728. [PMID: 31320905 PMCID: PMC6610765 DOI: 10.1155/2019/2186728] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/11/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) represent alternative candidates to chondrocytes for cartilage engineering. However, it remains difficult to identify the ideal source of MSCs for cartilage repair since conditions supporting chondrogenic induction are diverse among published works. In this study, we characterized and evaluated the chondrogenic potential of MSCs from bone marrow (BM), Wharton's jelly (WJ), dental pulp (DP), and adipose tissue (AT) isolated and cultivated under serum-free conditions. BM-, WJ-, DP-, and AT-MSCs did not differ in terms of viability, clonogenicity, and proliferation. By an extensive polychromatic flow cytometry analysis, we found notable differences in markers of the osteochondrogenic lineage between the 4 MSC sources. We then evaluated their chondrogenic potential in a micromass culture model, and only BM-MSCs showed chondrogenic conversion. This chondrogenic differentiation was specifically ascertained by the production of procollagen IIB, the only type II collagen isoform synthesized by well-differentiated chondrocytes. As a pilot study toward cartilage engineering, we encapsulated BM-MSCs in hydrogel and developed an original method to evaluate their chondrogenic conversion by flow cytometry analysis, after release of the cells from the hydrogel. This allowed the simultaneous quantification of procollagen IIB and α10, a subunit of a type II collagen receptor crucial for proper cartilage development. This work represents the first comparison of detailed immunophenotypic analysis and chondrogenic differentiation potential of human BM-, WJ-, DP-, and AT-MSCs performed under the same serum-free conditions, from their isolation to their induction. Our study, achieved in conditions compliant with clinical applications, highlights that BM-MSCs are good candidates for cartilage engineering.
Collapse
|
30
|
Takahashi H, Sakata N, Yoshimatsu G, Hasegawa S, Kodama S. Regenerative and Transplantation Medicine: Cellular Therapy Using Adipose Tissue-Derived Mesenchymal Stromal Cells for Type 1 Diabetes Mellitus. J Clin Med 2019; 8:jcm8020249. [PMID: 30781427 PMCID: PMC6406504 DOI: 10.3390/jcm8020249] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/09/2019] [Accepted: 02/13/2019] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the autoimmune targeting of pancreatic β-cells, and, in the advanced stage, severe hypoinsulinemia due to islet destruction. In patients with T1DM, continuous exogenous insulin therapy cannot be avoided. However, an insufficient dose of insulin easily induces extreme hyperglycemia or diabetic ketoacidosis, and intensive insulin therapy may cause hypoglycemic symptoms including hypoglycemic shock. While these insulin therapies are efficacious in most patients, some additional therapies are warranted to support the control of blood glucose levels and reduce the risk of hypoglycemia in patients who respond poorly despite receiving appropriate treatment. There has been a recent gain in the popularity of cellular therapies using mesenchymal stromal cells (MSCs) in various clinical fields, owing to their multipotentiality, capacity for self-renewal, and regenerative and immunomodulatory potential. In particular, adipose tissue-derived MSCs (ADMSCs) have become a focus in the clinical setting due to the abundance and easy isolation of these cells. In this review, we outline the possible therapeutic benefits of ADMSC for the treatment of T1DM.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Naoaki Sakata
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Gumpei Yoshimatsu
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Suguru Hasegawa
- Department of Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shohta Kodama
- Department of Regenerative Medicine & Transplantation, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
- Center for Regenerative Medicine, Fukuoka University Hospital, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| |
Collapse
|
31
|
Herten M, Zilkens C, Thorey F, Tassemeier T, Lensing-Höhn S, Fischer JC, Sager M, Krauspe R, Jäger M. Biomechanical Stability and Osteogenesis in a Tibial Bone Defect Treated by Autologous Ovine Cord Blood Cells-A Pilot Study. Molecules 2019; 24:molecules24020295. [PMID: 30650584 PMCID: PMC6358876 DOI: 10.3390/molecules24020295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to elucidate the impact of autologous umbilical cord blood cells (USSC) on bone regeneration and biomechanical stability in an ovine tibial bone defect. Ovine USSC were harvested and characterized. After 12 months, full-size 2.0 cm mid-diaphyseal bone defects were created and stabilized by an external fixateur containing a rigidity measuring device. Defects were filled with (i) autologous USSC on hydroxyapatite (HA) scaffold (test group), (ii) HA scaffold without cells (HA group), or (iii) left empty (control group). Biomechanical measures, standardized X-rays, and systemic response controls were performed regularly. After six months, bone regeneration was evaluated histomorphometrically and labeled USSC were tracked. In all groups, the torsion distance decreased over time, and radiographies showed comparable bone regeneration. The area of newly formed bone was 82.5 ± 5.5% in the control compared to 59.2 ± 13.0% in the test and 48.6 ± 2.9% in the HA group. Labeled cells could be detected in lymph nodes, liver and pancreas without any signs of tumor formation. Although biomechanical stability was reached earliest in the test group with autologous USSC on HA scaffold, the density of newly formed bone was superior in the control group without any bovine HA.
Collapse
Affiliation(s)
- Monika Herten
- Department of Orthopedics and Trauma Surgery, University of Duisburg-Essen, 45147 Essen, Germany.
| | - Christoph Zilkens
- Orthopedic Department, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Fritz Thorey
- Center for Hip, Knee and Foot Surgery, Sports Traumatology Department, ATOS Hospital, 69115 Heidelberg, Germany.
| | - Tjark Tassemeier
- Department of Orthopedics and Trauma Surgery, University of Duisburg-Essen, 45147 Essen, Germany.
| | - Sabine Lensing-Höhn
- Orthopedic Department, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Johannes C Fischer
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Martin Sager
- Animal Research Institute, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Rüdiger Krauspe
- Orthopedic Department, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany.
| | - Marcus Jäger
- Department of Orthopedics and Trauma Surgery, University of Duisburg-Essen, 45147 Essen, Germany.
| |
Collapse
|
32
|
Immunomodulatory Functions of Mesenchymal Stem Cells in Tissue Engineering. Stem Cells Int 2019; 2019:9671206. [PMID: 30766609 PMCID: PMC6350611 DOI: 10.1155/2019/9671206] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/26/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023] Open
Abstract
The inflammatory response to chronic injury affects tissue regeneration and has become an important factor influencing the prognosis of patients. In previous stem cell treatments, it was revealed that stem cells not only have the ability for direct differentiation or regeneration in chronic tissue damage but also have a regulatory effect on the immune microenvironment. Stem cells can regulate the immune microenvironment during tissue repair and provide a good "soil" for tissue regeneration. In the current study, the regulation of immune cells by mesenchymal stem cells (MSCs) in the local tissue microenvironment and the tissue damage repair mechanisms are revealed. The application of the concepts of "seed" and "soil" has opened up new research avenues for regenerative medicine. Tissue engineering (TE) technology has been used in multiple tissues and organs using its biomimetic and cellular cell abilities, and scaffolds are now seen as an important part of building seed cell microenvironments. The effect of tissue engineering techniques on stem cell immune regulation is related to the shape and structure of the scaffold, the preinflammatory microenvironment constructed by the implanted scaffold, and the material selection of the scaffold. In the application of scaffold, stem cell technology has important applications in cartilage, bone, heart, and liver and other research fields. In this review, we separately explore the mechanism of MSCs in different tissue and organs through immunoregulation for tissue regeneration and MSC combined with 3D scaffolds to promote MSC immunoregulation to repair damaged tissues.
Collapse
|
33
|
Longoni A, Knežević L, Schepers K, Weinans H, Rosenberg AJWP, Gawlitta D. The impact of immune response on endochondral bone regeneration. NPJ Regen Med 2018; 3:22. [PMID: 30510772 PMCID: PMC6265275 DOI: 10.1038/s41536-018-0060-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/26/2018] [Indexed: 12/29/2022] Open
Abstract
Tissue engineered cartilage substitutes, which induce the process of endochondral ossification, represent a regenerative strategy for bone defect healing. Such constructs typically consist of multipotent mesenchymal stromal cells (MSCs) forming a cartilage template in vitro, which can be implanted to stimulate bone formation in vivo. The use of MSCs of allogeneic origin could potentially improve the clinical utility of the tissue engineered cartilage constructs in three ways. First, ready-to-use construct availability can speed up the treatment process. Second, MSCs derived and expanded from a single donor could be applied to treat several patients and thus the costs of the medical interventions would decrease. Finally, it would allow more control over the quality of the MSC chondrogenic differentiation. However, even though the envisaged clinical use of allogeneic cell sources for bone regeneration is advantageous, their immunogenicity poses a significant obstacle to their clinical application. The aim of this review is to increase the awareness of the role played by immune cells during endochondral ossification, and in particular during regenerative strategies when the immune response is altered by the presence of implanted biomaterials and/or cells. More specifically, we focus on how this balance between immune response and bone regeneration is affected by the implantation of a cartilaginous tissue engineered construct of allogeneic origin.
Collapse
Affiliation(s)
- A Longoni
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| | - L Knežević
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,3Faculty of Health Sciences, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD UK
| | - K Schepers
- 4Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - H Weinans
- 5Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, 3508 GA Utrecht, The Netherlands.,6Department of Rheumatology, University Medical Center Utrecht, Utrecht University, 3584CX Utrecht, The Netherlands.,7Department of Biomechanical Engineering, Delft University of Technology, 2628CD Delft, The Netherlands
| | - A J W P Rosenberg
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands
| | - D Gawlitta
- 1Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, G05.222, PO Box 85500, 3508 GA The Netherlands.,Regenerative Medicine Center Utrecht, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
34
|
Schulman IH, Balkan W, Hare JM. Mesenchymal Stem Cell Therapy for Aging Frailty. Front Nutr 2018; 5:108. [PMID: 30498696 PMCID: PMC6249304 DOI: 10.3389/fnut.2018.00108] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022] Open
Abstract
Chronic diseases and degenerative conditions are strongly linked with the geriatric syndrome of frailty and account for a disproportionate percentage of the health care budget. Frailty increases the risk of falls, hospitalization, institutionalization, disability, and death. By definition, frailty syndrome is characterized by declines in lean body mass, strength, endurance, balance, gait speed, activity and energy levels, and organ physiologic reserve. Collectively, these changes lead to the loss of homeostasis and capability to withstand stressors and resulting vulnerabilities. There is a strong link between frailty, inflammation, and the impaired ability to repair tissue injury due to decreases in endogenous stem cell production. Although exercise and nutritional supplementation provide benefit to frail patients, there are currently no specific therapies for frailty. Bone marrow-derived allogeneic mesenchymal stem cells (MSCs) provide therapeutic benefits in heart failure patients irrespective of age. MSCs contribute to cellular repair and tissue regeneration through their multilineage differentiation capacity, immunomodulatory, and anti-inflammatory effects, homing and migratory capacity to injury sites, and stimulatory effect on endogenous tissue progenitors. The advantages of using MSCs as a therapeutic strategy include standardization of isolation and culture expansion techniques and safety in allogeneic transplantation. Based on this evidence, we performed a randomized, double-blinded, dose-finding study in elderly, frail individuals and showed that intravenously delivered allogeneic MSCs are safe and produce significant improvements in physical performance measures and inflammatory biomarkers. We thus propose that frailty can be treated and the link between frailty and chronic inflammation offers a potential therapeutic target, addressable by cell therapy.
Collapse
Affiliation(s)
- Ivonne Hernandez Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States.,Katz Family Division of Nephrology and Hypertension, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
35
|
|
36
|
White JL, Walker NJ, Hu JC, Borjesson DL, Athanasiou KA. A Comparison of Bone Marrow and Cord Blood Mesenchymal Stem Cells for Cartilage Self-Assembly. Tissue Eng Part A 2018; 24:1262-1272. [PMID: 29478385 DOI: 10.1089/ten.tea.2017.0424] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Joint injury is a common cause of premature retirement for the human and equine athlete alike. Implantation of engineered cartilage offers the potential to increase the success rate of surgical intervention and hasten recovery times. Mesenchymal stem cells (MSCs) are a particularly attractive cell source for cartilage engineering. While bone marrow-derived MSCs (BM-MSCs) have been most extensively characterized for musculoskeletal tissue engineering, studies suggest that cord blood MSCs (CB-MSCs) may elicit a more robust chondrogenic phenotype. The objective of this study was to determine a superior equine MSC source for cartilage engineering. MSCs derived from bone marrow or cord blood were stimulated to undergo chondrogenesis through aggregate redifferentiation and used to generate cartilage through the self-assembling process. The resulting neocartilage produced from either BM-MSCs or CB-MSCs was compared by measuring mechanical, biochemical, and histological properties. We found that while BM constructs possessed higher tensile properties and collagen content, CB constructs had superior compressive properties comparable to that of native tissue and higher GAG content. Moreover, CB constructs had alkaline phosphatase activity, collagen type X, and collagen type II on par with native tissue suggesting a more hyaline cartilage-like phenotype. In conclusion, while both BM-MSCs and CB-MSCs were able to form neocartilage, CB-MSCs resulted in tissue more closely resembling native equine articular cartilage as determined by a quantitative functionality index. Therefore, CB-MSCs are deemed a superior source for the purpose of articular cartilage self-assembly.
Collapse
Affiliation(s)
- Jamie L White
- 1 Department of Pathology, Microbiology and Immunology, Integrative Pathobiology Graduate Group, University of California , Davis, Davis, California
| | - Naomi J Walker
- 2 Department of Pathology, Microbiology and Immunology, University of California , Davis, Davis, California
| | - Jerry C Hu
- 3 Department of Biomedical Engineering, Henry Samueli School of Engineering, University of California , Irvine, Irvine, California
| | - Dori L Borjesson
- 2 Department of Pathology, Microbiology and Immunology, University of California , Davis, Davis, California.,4 School of Veterinary Medicine, Veterinary Institute for Regenerative Cures, University of California , Davis, Davis, California
| | - Kyriacos A Athanasiou
- 3 Department of Biomedical Engineering, Henry Samueli School of Engineering, University of California , Irvine, Irvine, California
| |
Collapse
|
37
|
Bedian L, Villalba-Rodríguez AM, Hernández-Vargas G, Parra-Saldivar R, Iqbal HMN. Bio-based materials with novel characteristics for tissue engineering applications - A review. Int J Biol Macromol 2017; 98:837-846. [PMID: 28223133 DOI: 10.1016/j.ijbiomac.2017.02.048] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 02/05/2023]
Abstract
Recently, a wider spectrum of bio-based materials and materials-based novel constructs and systems has been engineered with high interests. The key objective is to help for an enhanced/better quality of life in a secure way by avoiding/limiting various adverse effects of some in practice traditional therapies. In this context, different methodological approaches including in vitro, in vivo, and ex vivo techniques have been exploited, so far. Among them, bio-based therapeutic constructs are of supreme interests for an enhanced and efficient delivery in the current biomedical sector of the modern world. The development of new types of novel, effective and highly reliable materials-based novel constructs for multipurpose applications is essential and a core demand to tackle many human health related diseases. Bio-based materials possess several complementary functionalities, e.g. unique chemical structure, bioactivity, non-toxicity, biocompatibility, biodegradability, recyclability, etc. that position them well in the modern world's materials sector. In this context, the utilization of biomaterials provides extensive opportunities for experimentation in the field of interdisciplinary and multidisciplinary scientific research. With an aim to address the global dependence on petroleum-based polymers, researchers have been redirecting their interests to the engineering of biological materials for targeted applications in different industries including cosmetics, pharmaceuticals, and other biotechnological or biomedical applications. Herein, we reviewed biotechnological advancements at large and tissue engineering from a biomaterials perspective in particular and envision directions of future developments.
Collapse
Affiliation(s)
- Luis Bedian
- School of Engineering and Science, Tecnologico de Monterrey, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. CP 64849, Mexico
| | - Angel M Villalba-Rodríguez
- School of Engineering and Science, Tecnologico de Monterrey, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. CP 64849, Mexico
| | - Gustavo Hernández-Vargas
- School of Engineering and Science, Tecnologico de Monterrey, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. CP 64849, Mexico
| | - Roberto Parra-Saldivar
- School of Engineering and Science, Tecnologico de Monterrey, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. CP 64849, Mexico
| | - Hafiz M N Iqbal
- School of Engineering and Science, Tecnologico de Monterrey, Campus Monterrey, Ave. Eugenio Garza Sada 2501, Monterrey, N.L. CP 64849, Mexico.
| |
Collapse
|
38
|
Koh RH, Jin Y, Kang BJ, Hwang NS. Chondrogenically primed tonsil-derived mesenchymal stem cells encapsulated in riboflavin-induced photocrosslinking collagen-hyaluronic acid hydrogel for meniscus tissue repairs. Acta Biomater 2017; 53:318-328. [PMID: 28161573 DOI: 10.1016/j.actbio.2017.01.081] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/26/2017] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Current meniscus tissue repairing strategies involve partial or total meniscectomy, followed by allograft transplantation or synthetic material implantation. However, allografts and synthetic implants have major drawbacks such as the limited supply of grafts and lack of integration into host tissue, respectively. In this study, we investigated the effects of conditioned medium (CM) from meniscal fibrochondrocytes and TGF-β3 on tonsil-derived mesenchymal stem cells (T-MSCs) for meniscus tissue engineering. CM-expanded T-MSCs were encapsulated in riboflavin-induced photocrosslinked collagen-hyaluronic acid (COL-RF-HA) hydrogels and cultured in chondrogenic medium containing TGF-β3. In vitro results indicate that CM-expanded cells followed by TGF-β3 exposure stimulated the expression of fibrocartilage-related genes (COL2, SOX9, ACAN, COL1) and production of extracellular matrix components. Histological assessment of in vitro and subcutaneously implanted in vivo constructs demonstrated that CM-expanded cells followed by TGF-β3 exposure resulted in highest cell proliferation, GAG accumulation, and collagen deposition. Furthermore, when implanted into meniscus defect model, CM treatment amplified the potential of TGF-β3 and induced complete regeneration. STATEMENT OF SIGNIFICANCE Conditioned medium derived from chondrocytes have been reported to effectively prime mesenchymal stem cells toward chondrogenic lineage. Type I collagen is the main component of meniscus extracellular matrix and hyaluronic acid is known to promote meniscus regeneration. In this manuscript, we investigated the effects of conditioned medium (CM) and transforming growth factor-β3 (TGF-β3) on tonsil-derived mesenchymal stem cells (T-MSCs) encapsulated in riboflavin-induced photocrosslinked collagen-hyaluronic acid (COL-RF-HA) hydrogel. We employed a novel source of conditioned medium, derived from meniscal fibrochondrocytes. Our in vitro and in vivo results collectively illustrate that CM-expanded cells followed by TGF-β3 exposure have the best potential for meniscus regeneration. This manuscript highlights a novel stem cell commitment strategy combined with biomaterials designs for meniscus regeneration.
Collapse
|
39
|
Yu H, Cauchois G, Schmitt JF, Louvet N, Six JL, Chen Y, Rahouadj R, Huselstein C. Is there a cause-and-effect relationship between physicochemical properties and cell behavior of alginate-based hydrogel obtained after sterilization? J Mech Behav Biomed Mater 2017; 68:134-143. [DOI: 10.1016/j.jmbbm.2017.01.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/21/2017] [Accepted: 01/24/2017] [Indexed: 11/25/2022]
|
40
|
Guerado E, Caso E. Challenges of bone tissue engineering in orthopaedic patients. World J Orthop 2017; 8:87-98. [PMID: 28251059 PMCID: PMC5314152 DOI: 10.5312/wjo.v8.i2.87] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/23/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023] Open
Abstract
Bone defects may impede normal biomechanics and the structural stability of bone as an organ. In many cases, the correction of bone defects requires extensive surgical intervention involving the use of bone-grafting techniques and other procedures in which healing is slow, there is a high risk of infection and considerable pain is provoked - with no guarantee of complete correction of the defect. Therefore, the search for surgical alternatives continues to present a major challenge in orthopaedic traumatology. The reamer-irrigator-aspirator (RIA) system, which was devised to avoid the problems that can arise with autograft harvesting from the iliac crest, consists of collecting the product of the femoral canal after reaming. The RIA technique improves osteogenic differentiation of mesenchymal stem cells, compared to bone marrow aspiration or cancellous bone harvesting from the iliac crest using a spoon. Another approach, the Masquelet technique, consists of reconstructing a long bone defect by means of an induced membrane grown onto an acrylic cement rod inserted to fill the defect; in a second surgical step, once the membrane is constituted, the cement rod is removed and cancellous autograft is used to fill the defect. Both in RIA and in the Masquelet technique, osteosynthesis is usually needed. Bone transportation by compression-distraction lengthening principles is commonly implemented for the treatment of large bone loss. However, complications are frequently encountered with these techniques. Among new techniques that have been proposed to address the problem of large bone loss, the application of stem cells in conjunction with tissue engineering techniques is very promising, as is the creation of personalised medicine (or precision medicine), in which molecular profiling technologies are used to tailor the therapeutic strategy, to ensure the right method is applied for the right person at the right time, after determining the predisposition to disease among the general population. All of the above techniques for addressing bone defects are discussed in this paper.
Collapse
|