1
|
Yuan Y, Liu T. Influence of mesenchymal stem cells from different origins on the therapeutic effectiveness of systemic lupus erythematosus. Exp Cell Res 2024; 442:114263. [PMID: 39307406 DOI: 10.1016/j.yexcr.2024.114263] [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: 06/17/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
Systemic Lupus Erythematosus (SLE) is a chronic autoimmune inflammatory disorder characterized by alterations in the balance between inflammatory and regulatory cytokines. Mesenchymal stem cells (MSCs), which are non-hematopoietic stem cells with multipotent differentiation potential, due to their immunomodulatory, tissue repair, low immunogenicity, and chemotactic properties, have garnered increasing interest in SLE treatment. Studies increasingly reveal the heterogeneous nature of MSC populations. With sources including dental pulp, adipose tissue, bone marrow, and umbilical cord, the therapeutic effects of MSCs on SLE vary depending on their origin. This review consolidates clinical research on MSCs from different sources in treating SLE and analyzes the possible causes underlying these variable outcomes. Additionally, it elucidates five potential factors impacting the outcomes of MSC therapy in SLE: the influence of the microenvironment on MSCs, the complexity and paradoxical aspects of MSC mechanisms in SLE treatment, the heterogeneity of MSCs, the in vivo differentiation potential and post-transplant survival rates of MSCs, and disparities in MSC preparation conditions.
Collapse
Affiliation(s)
- Yuan Yuan
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China.
| | - Tong Liu
- Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, China
| |
Collapse
|
2
|
Moldaschl J, Chariyev-Prinz F, Toegel S, Keck M, Hiden U, Egger D, Kasper C. Spheroid trilineage differentiation model of primary mesenchymal stem/stromal cells under hypoxia and serum-free culture conditions. Front Bioeng Biotechnol 2024; 12:1444363. [PMID: 39144480 PMCID: PMC11321963 DOI: 10.3389/fbioe.2024.1444363] [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: 06/05/2024] [Accepted: 07/12/2024] [Indexed: 08/16/2024] Open
Abstract
Due to their unique properties, human mesenchymal stem/stromal cells (MSCs) possess tremendous potential in regenerative medicine, particularly in cell-based therapies where the multipotency and immunomodulatory characteristics of MSCs can be leveraged to address a variety of disease states. Although MSC-based cell therapeutics have emerged as one of the most promising medical treatments, the clinical translation is hampered by the variability of MSC-based cellular products caused by tissue source-specific differences and the lack of physiological cell culture approaches that closely mimic the human cellular microenvironment. In this study, a model for trilineage differentiation of primary adipose-, bone marrow-, and umbilical cord-derived MSCs into adipocytes, chondrocytes and osteoblasts was established and characterized. Differentiation was performed in spheroid culture, using hypoxic conditions and serum-free and antibiotics-free medium. This platform was characterized for spheroid diameter and trilineage differentiation capacity reflecting functionality of differentiated cells, as indicated by lineage-specific extracellular matrix (ECM) accumulation and expression of distinct secreted markers. The presented model shows spheroid growth during the course of differentiation and successfully supports trilineage differentiation for MSCs from almost all tissue sources except for osteogenesis of umbilical cord-derived MSCs. These findings indicate that this platform provides a suitable and favorable environment for trilineage differentiation of MSCs from various tissue sources. Therefore, it poses a promising model to generate highly relevant biological data urgently required for clinical translation and therefore might be used in the future to generate in vitro microtissues, building blocks for tissue engineering or as disease models.
Collapse
Affiliation(s)
- Julia Moldaschl
- Institute of Cell and Tissue Culture Technologies, BOKU University, Vienna, Austria
| | | | - Stefan Toegel
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Maike Keck
- Department of Plastic, Reconstructive and Aesthetic Surgery, Agaplesion Diakonieklinikum Hamburg, Hamburg, Germany
- Klinik für Plastische Chirurgie, Universität zu Lübeck, Lübeck, Germany
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Dominik Egger
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany
| | - Cornelia Kasper
- Institute of Cell and Tissue Culture Technologies, BOKU University, Vienna, Austria
| |
Collapse
|
3
|
Otani Y, Schol J, Sakai D, Nakamura Y, Sako K, Warita T, Tamagawa S, Ambrosio L, Munesada D, Ogasawara S, Matsushita E, Kawachi A, Naiki M, Sato M, Watanabe M. Assessment of Tie2-Rejuvenated Nucleus Pulposus Cell Transplants from Young and Old Patient Sources Demonstrates That Age Still Matters. Int J Mol Sci 2024; 25:8335. [PMID: 39125917 PMCID: PMC11312270 DOI: 10.3390/ijms25158335] [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: 05/13/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Cell transplantation is being actively explored as a regenerative therapy for discogenic back pain. This study explored the regenerative potential of Tie2+ nucleus pulposus progenitor cells (NPPCs) from intervertebral disc (IVD) tissues derived from young (<25 years of age) and old (>60 years of age) patient donors. We employed an optimized culture method to maintain Tie2 expression in NP cells from both donor categories. Our study revealed similar Tie2 positivity rates regardless of donor types following cell culture. Nevertheless, clear differences were also found, such as the emergence of significantly higher (3.6-fold) GD2 positivity and reduced (2.7-fold) proliferation potential for older donors compared to young sources. Our results suggest that, despite obtaining a high fraction of Tie2+ NP cells, cells from older donors were already committed to a more mature phenotype. These disparities translated into functional differences, influencing colony formation, extracellular matrix production, and in vivo regenerative potential. This study underscores the importance of considering age-related factors in NPPC-based therapies for disc degeneration. Further investigation into the genetic and epigenetic alterations of Tie2+ NP cells from older donors is crucial for refining regenerative strategies. These findings shed light on Tie2+ NPPCs as a promising cell source for IVD regeneration while emphasizing the need for comprehensive understanding and scalability considerations in culture methods for broader clinical applicability.
Collapse
Affiliation(s)
- Yuto Otani
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
| | - Jordy Schol
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Center for Musculoskeletal Innovative Research and Advancement (C-MiRA), Tokai University Graduate School, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Daisuke Sakai
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Center for Musculoskeletal Innovative Research and Advancement (C-MiRA), Tokai University Graduate School, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Yoshihiko Nakamura
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
| | - Kosuke Sako
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
| | - Takayuki Warita
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- TUNZ Pharma Corporation, Osaka 541-0046, Japan;
| | - Shota Tamagawa
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Luca Ambrosio
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 01128 Rome, Italy
| | - Daiki Munesada
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
| | - Shota Ogasawara
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
| | - Erika Matsushita
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Center for Musculoskeletal Innovative Research and Advancement (C-MiRA), Tokai University Graduate School, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Asami Kawachi
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- TUNZ Pharma Corporation, Osaka 541-0046, Japan;
| | | | - Masato Sato
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Center for Musculoskeletal Innovative Research and Advancement (C-MiRA), Tokai University Graduate School, 143 Shimokasuya, Isehara 259-1193, Japan
| | - Masahiko Watanabe
- Department of Orthopedic Surgery, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (Y.O.); (J.S.); (Y.N.); (K.S.); (T.W.); (S.T.); (L.A.); (D.M.); (S.O.); (A.K.); (M.S.); (M.W.)
- Center for Musculoskeletal Innovative Research and Advancement (C-MiRA), Tokai University Graduate School, 143 Shimokasuya, Isehara 259-1193, Japan
| |
Collapse
|
4
|
Kondratenko AA, Tovpeko DV, Volov DA, Kalyuzhnaya LI, Chernov VE, Glushakov RI, Sirotkina MY, Zemlyanoy DA, Bildyug NB, Chebotarev SV, Alexander-Sinclair EI, Nashchekin AV, Belova AD, Grigoriev AM, Kirsanova LA, Basok YB, Sevastianov VI. Decellularized Umbilical Cord as a Scaffold to Support Healing of Full-Thickness Wounds. Biomimetics (Basel) 2024; 9:405. [PMID: 39056846 PMCID: PMC11274938 DOI: 10.3390/biomimetics9070405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
The umbilical cord is a material that enhances regeneration and is devoid of age-related changes in the extracellular matrix (ECM). The aim of this work was to develop a biodegradable scaffold from a decellularized human umbilical cord (UC-scaffold) to heal full-thickness wounds. Decellularization was performed with 0.05% sodium dodecyl sulfate solution. The UC-scaffold was studied using morphological analysis methods. The composition of the UC-scaffold was studied using immunoblotting and Fourier transform infrared spectroscopy. The adhesion and proliferation of mesenchymal stromal cells were investigated using the LIVE/DEAD assay. The local reaction was determined by subcutaneous implantation in mice (n = 60). A model of a full-thickness skin wound in mice (n = 64) was used to assess the biological activity of the UC-scaffold. The proposed decellularization method showed its effectiveness in the umbilical cord, as it removed cells and retained a porous structure, type I and type IV collagen, TGF-β3, VEGF, and fibronectin in the ECM. The biodegradation of the UC-scaffold in the presence of collagenase, its stability during incubation in hyaluronidase solution, and its ability to swell by 1617 ± 120% were demonstrated. Subcutaneous scaffold implantation in mice showed gradual resorption of the product in vivo without the formation of a dense connective tissue capsule. Epithelialization of the wound occurred completely in contrast to the controls. All of these data suggest a potential for the use of the UC-scaffold.
Collapse
Affiliation(s)
- Albina A. Kondratenko
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
- Department of Histology and Embryology, St. Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Dmitry V. Tovpeko
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Daniil A. Volov
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Lidia I. Kalyuzhnaya
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Vladimir E. Chernov
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Ruslan I. Glushakov
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
- Department of Pharmacology with a Course of Clinical Pharmacology and Pharmacoeconomics, St. Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Maria Y. Sirotkina
- Cellular biotechnology Centre for Cell Technology (CCT), Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia (N.B.B.)
| | - Dmitry A. Zemlyanoy
- Department of General Hygiene, St. Petersburg State Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Natalya B. Bildyug
- Cellular biotechnology Centre for Cell Technology (CCT), Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia (N.B.B.)
| | - Sergey V. Chebotarev
- Research Department of Biomedical Research of the Research Center, S.M. Kirov Military Medical Academy, 194044 St. Petersburg, Russia
| | - Elga I. Alexander-Sinclair
- Cellular biotechnology Centre for Cell Technology (CCT), Institute of Cytology of the Russian Academy of Sciences, 194064 St. Petersburg, Russia (N.B.B.)
| | - Alexey V. Nashchekin
- Laboratory “Characterization of Materials and Structures of Solid State Electronics”, Ioffe Institute, 194021 St. Petersburg, Russia
| | - Aleksandra D. Belova
- Department for Biomedical Technologies and Tissue Engineering. Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia
| | - Alexey M. Grigoriev
- Department for Biomedical Technologies and Tissue Engineering. Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia
| | - Ludmila A. Kirsanova
- Department for Biomedical Technologies and Tissue Engineering. Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia
| | - Yulia B. Basok
- Department for Biomedical Technologies and Tissue Engineering. Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia
| | - Victor I. Sevastianov
- Department for Biomedical Technologies and Tissue Engineering. Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia
| |
Collapse
|
5
|
Scognamiglio F, Pizzolitto C, Romano M, Teti G, Zara S, Conz M, Donati I, Porrelli D, Falconi M, Marsich E. "A lactose-modified chitosan accelerates chondrogenic differentiation in mesenchymal stem cells spheroids". BIOMATERIALS ADVANCES 2024; 160:213849. [PMID: 38599041 DOI: 10.1016/j.bioadv.2024.213849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
Spheroids derived from human mesenchymal stem cells (hMSCs) are of limited use for cartilage regeneration, as the viability of the cells progressively decreases during the period required for chondrogenic differentiation (21 days). In this work, spheroids based on hMSCs and a lactose-modified chitosan (CTL) were formed by seeding cells onto an air-dried coating of CTL. The polymer coating can inhibit cell adhesion and it is simultaneously incorporated into spheroid structure. CTL-spheroids were characterized from a morphological and biological perspective, and their properties were compared with those of spheroids obtained by seeding the cells onto a non-adherent surface (agar gel). Compared to the latter, smaller and more viable spheroids form in the presence of CTL as early as 4 days of culture. At this time point, analysis of stem cells differentiation in spheroids showed a remarkable increase in collagen type-2 (COL2A1) gene expression (~700-fold compared to day 0), whereas only a 2-fold increase was observed in the control spheroids at day 21. These results were confirmed by histological and transmission electron microscopy (TEM) analyses, which showed that in CTL-spheroids an early deposition of collagen with a banding structure already occurred at day 7. Overall, these results support the use of CTL-spheroids as a novel system for cartilage regeneration, characterized by increased cell viability and differentiation capacity within a short time-frame. This will pave the way for approaches aimed at increasing the success rate of procedures and reducing the time required for tissue regeneration.
Collapse
Affiliation(s)
- F Scognamiglio
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy.
| | - C Pizzolitto
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy.
| | - M Romano
- Department of Life Sciences, University of Trieste, Via Valerio 28, 34127 Trieste, Italy.
| | - G Teti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - S Zara
- Department of Pharmacy, University "G. d'Annunzio" Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy.
| | - M Conz
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy.
| | - I Donati
- Department of Life Sciences, University of Trieste, Via Licio Giorgieri 5, 34127 Trieste, Italy.
| | - D Porrelli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy.
| | - M Falconi
- Department of Medical and Surgical Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - E Marsich
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy.
| |
Collapse
|
6
|
Nordberg RC, Bielajew BJ, Takahashi T, Dai S, Hu JC, Athanasiou KA. Recent advancements in cartilage tissue engineering innovation and translation. Nat Rev Rheumatol 2024; 20:323-346. [PMID: 38740860 PMCID: PMC11524031 DOI: 10.1038/s41584-024-01118-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 05/16/2024]
Abstract
Articular cartilage was expected to be one of the first successfully engineered tissues, but today, cartilage repair products are few and they exhibit considerable limitations. For example, of the cell-based products that are available globally, only one is marketed for non-knee indications, none are indicated for severe osteoarthritis or rheumatoid arthritis, and only one is approved for marketing in the USA. However, advances in cartilage tissue engineering might now finally lead to the development of new cartilage repair products. To understand the potential in this field, it helps to consider the current landscape of tissue-engineered products for articular cartilage repair and particularly cell-based therapies. Advances relating to cell sources, bioactive stimuli and scaffold or scaffold-free approaches should now contribute to progress in therapeutic development. Engineering for an inflammatory environment is required because of the need for implants to withstand immune challenge within joints affected by osteoarthritis or rheumatoid arthritis. Bringing additional cartilage repair products to the market will require an understanding of the translational vector for their commercialization. Advances thus far can facilitate the future translation of engineered cartilage products to benefit the millions of patients who suffer from cartilage injuries and arthritides.
Collapse
Affiliation(s)
- Rachel C Nordberg
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Benjamin J Bielajew
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Takumi Takahashi
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Shuyan Dai
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
7
|
Vakhrushev IV, Basok YB, Baskaev KK, Novikova VD, Leonov GE, Grigoriev AM, Belova AD, Kirsanova LA, Lupatov AY, Burunova VV, Kovalev AV, Makarevich PI, Sevastianov VI, Yarygin KN. Cartilage-Specific Gene Expression and Extracellular Matrix Deposition in the Course of Mesenchymal Stromal Cell Chondrogenic Differentiation in 3D Spheroid Culture. Int J Mol Sci 2024; 25:5695. [PMID: 38891883 PMCID: PMC11172056 DOI: 10.3390/ijms25115695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Articular cartilage damage still remains a major problem in orthopedical surgery. The development of tissue engineering techniques such as autologous chondrocyte implantation is a promising way to improve clinical outcomes. On the other hand, the clinical application of autologous chondrocytes has considerable limitations. Mesenchymal stromal cells (MSCs) from various tissues have been shown to possess chondrogenic differentiation potential, although to different degrees. In the present study, we assessed the alterations in chondrogenesis-related gene transcription rates and extracellular matrix deposition levels before and after the chondrogenic differentiation of MSCs in a 3D spheroid culture. MSCs were obtained from three different tissues: umbilical cord Wharton's jelly (WJMSC-Wharton's jelly mesenchymal stromal cells), adipose tissue (ATMSC-adipose tissue mesenchymal stromal cells), and the dental pulp of deciduous teeth (SHEDs-stem cells from human exfoliated deciduous teeth). Monolayer MSC cultures served as baseline controls. Newly formed 3D spheroids composed of MSCs previously grown in 2D cultures were precultured for 2 days in growth medium, and then, chondrogenic differentiation was induced by maintaining them in the TGF-β1-containing medium for 21 days. Among the MSC types studied, WJMSCs showed the most similarities with primary chondrocytes in terms of the upregulation of cartilage-specific gene expression. Interestingly, such upregulation occurred to some extent in all 3D spheroids, even prior to the addition of TGF-β1. These results confirm that the potential of Wharton's jelly is on par with adipose tissue as a valuable cell source for cartilage engineering applications as well as for the treatment of osteoarthritis. The 3D spheroid environment on its own acts as a trigger for the chondrogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Igor V. Vakhrushev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Yulia B. Basok
- Department for Biomedical Technologies and Tissue Engineering, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow 123182, Russia; (Y.B.B.); (A.M.G.); (A.D.B.); (L.A.K.); (V.I.S.)
| | - Konstantin K. Baskaev
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Victoria D. Novikova
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Georgy E. Leonov
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Alexey M. Grigoriev
- Department for Biomedical Technologies and Tissue Engineering, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow 123182, Russia; (Y.B.B.); (A.M.G.); (A.D.B.); (L.A.K.); (V.I.S.)
| | - Aleksandra D. Belova
- Department for Biomedical Technologies and Tissue Engineering, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow 123182, Russia; (Y.B.B.); (A.M.G.); (A.D.B.); (L.A.K.); (V.I.S.)
| | - Ludmila A. Kirsanova
- Department for Biomedical Technologies and Tissue Engineering, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow 123182, Russia; (Y.B.B.); (A.M.G.); (A.D.B.); (L.A.K.); (V.I.S.)
| | - Alexey Y. Lupatov
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Veronika V. Burunova
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| | - Alexey V. Kovalev
- Priorov Central Institute for Trauma and Orthopedics, Moscow 127299, Russia;
| | - Pavel I. Makarevich
- Institute for Regenerative Medicine, Medical Research and Education Centre, Lomonosov Moscow State University, Moscow 119192, Russia;
| | - Victor I. Sevastianov
- Department for Biomedical Technologies and Tissue Engineering, Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow 123182, Russia; (Y.B.B.); (A.M.G.); (A.D.B.); (L.A.K.); (V.I.S.)
- Institute of Biomedical Research and Technology, Moscow 123557, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, Institute of Biomedical Chemistry, Moscow 119121, Russia; (K.K.B.); (V.D.N.); (G.E.L.); (V.V.B.); (K.N.Y.)
| |
Collapse
|
8
|
Arki MK, Moeinabadi-Bidgoli K, Niknam B, Mohammadi P, Hassan M, Hossein-Khannazer N, Vosough M. Immunomodulatory performance of GMP-compliant, clinical-grade mesenchymal stromal cells from four different sources. Heliyon 2024; 10:e24948. [PMID: 38312681 PMCID: PMC10835001 DOI: 10.1016/j.heliyon.2024.e24948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/06/2024] Open
Abstract
Inflammatory and autoimmune diseases are among the most challenging disorders for health care professionals that require systemic immune suppression which associates with various side effects. Mesenchymal stromal cells (MSCs) are capable of regulating immune responses, mainly through paracrine effects and cell-cell contact. Since MSCs are advanced therapy medicinal products (ATMPs), they must follow Good Manufacturing Practice (GMP) regulations to ensure their safety and efficacy. In this study, we evaluated the immunomodulatory effects of GMP-compliant clinical grade MSCs obtained from four different sources (bone marrow, adipose tissue, Wharton's Jelly, and decidua tissue) on allogeneic peripheral blood mononuclear cells (PBMCs). Our results revealed that WJ-MSCs were the most successful group in inhibiting PBMC proliferation as confirmed by BrdU analysis. Moreover, WJ-MSCs were the strongest group in enhancing the regulatory T cell population of PBMCs. WJ-MSCs also had the highest secretory profile of prostaglandin E2 (PGE-2), anti-inflammatory cytokine, while interleukin-10 (IL-10) secretion was highest in the DS-MSC group. DS-MSCs also had the lowest secretion of IL-12 and IL-17 inflammatory cytokines. Transcriptome analysis revealed that WJ-MSCs had the lowest expression of IL-6, while DS-MSCs were the most potent group in the expression of immunomodulatory factors such as hepatocyte growth factor (HGF) and transforming growth factor-β (TGF- β). Taken together, our results indicated that GMP-compliant Wharton's Jelly and decidua-derived MSCs showed the best immunomodulatory performance considering paracrine factors.
Collapse
Affiliation(s)
- Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kasra Moeinabadi-Bidgoli
- Basic and Molecular Epidemiology of Gastroenterology Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Niknam
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parvaneh Mohammadi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83, Stockholm, Sweden
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 141-83, Stockholm, Sweden
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Ehioghae M, Vippa TK, Askins D, Slusarczyk S, Bobo E, Montoya A, Anderson D, Robinson CL, Kaye AD, Urits I. Exploring Orthopedic Stem-Cell Approaches for Osteoarthritis Management: Current Trends and Future Horizons. Curr Pain Headache Rep 2024; 28:27-35. [PMID: 38010488 DOI: 10.1007/s11916-023-01191-6] [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] [Accepted: 11/20/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE OF REVIEW Osteoarthritis (OA) is a prevalent and debilitating condition characterized by joint degeneration and pain. Current treatment options aim to alleviate symptoms and slow disease progression but lack curative potential. Stem cell therapies have emerged as a promising alternative. This article explores the epidemiology, pathophysiology, clinical manifestations of hip and knee OA, and the evolving role of stem cell therapies in their treatment. RECENT FINDINGS The global prevalence of OA, with knee OA being the most common form, has fueled the demand for stem cell therapies. Despite limited robust evidence supporting their efficacy, clinical trials investigating stem-cell treatments for OA have reported encouraging radiological and clinical improvements. Stem cell therapies offer potential disease-modifying benefits through immunomodulatory actions, growth factor secretion, and chondrogenic capabilities. Adipose-derived mesenchymal stem cells (ADMSCs) have shown promise in clinical trials for OA treatment, offering potential pain relief and functional improvement. ADMSCs possess advantages such as accessibility and a favorable safety profile, making them a viable option for OA management. Although other stem-cell types, including human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), have been used in OA treatment, ADMSCs have demonstrated superior outcomes. By providing a comprehensive overview of the evolving landscape of stem cell therapies for hip and knee OA, this article highlights the potential of stem-cell treatments to address the limitations of current therapies. However, further research is required to establish their long-term efficacy, identify optimal stem-cell types, and develop standardized protocols.
Collapse
Affiliation(s)
| | | | | | | | - Elena Bobo
- Tulane University School of Medicine, New Orleans, USA
| | - Alexis Montoya
- University of Arizona College of Medicine-Phoenix, Phoenix, USA
| | | | | | - Alan D Kaye
- Louisiana State University Health Shreveport, Shreveport, USA
| | | |
Collapse
|
10
|
Liu S. Scaffolded Chondrogenic Spheroid-Engrafted Model. Methods Mol Biol 2024; 2766:17-24. [PMID: 38270862 DOI: 10.1007/978-1-0716-3682-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
Therapeutic approaches using mesenchymal stem cells (MSCs) for a cartilage regeneration strategy are based on their multipotent differentiation for skeletal regeneration. With the utilization of allergenic neutralized type I atelocollagen during the pre-formation of chondrogenic MSC spheroids, cellular condensation and chondrogenic differentiation can be easily achieved. It also benefits the recruitment of host MSCs, which differentiate into chondrocyte-like cells after implantation into the experiment model. Using pre-formed chondrogenic MSC spheroids, the efficacy of anti-rheumatoid agents for cartilage repair can be screened on a large scale ex vivo. Furthermore, atelocollagen-scaffolded chondrogenic spheroids can be utilized for in vivo transplantation into a humanized xenografted arthritis model. Thus, the ability of cartilage self-repair can be qualitatively and quantitatively evaluated.
Collapse
Affiliation(s)
- Shuang Liu
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan.
| |
Collapse
|
11
|
Lee DH, Park KS, Shin HE, Kim SB, Choi H, An SB, Choi H, Kim JP, Han I. Safety and Feasibility of Intradiscal Administration of Matrilin-3-Primed Adipose-Derived Mesenchymal Stromal Cell Spheroids for Chronic Discogenic Low Back Pain: Phase 1 Clinical Trial. Int J Mol Sci 2023; 24:16827. [PMID: 38069151 PMCID: PMC10706656 DOI: 10.3390/ijms242316827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Functionally enhanced mesenchymal stromal cells participate in the repair of intervertebral disc. This study aimed to assess the safety and tolerability of intradiscal administration of matrilin-3-primed adipose-derived stromal cell (ASC) spheroids with hyaluronic acid (HA) in patients with chronic discogenic low back pain (LBP). In this single-arm, open-label phase I clinical trial, eight patients with chronic discogenic LBP were observed over 6 months. Each patient underwent a one-time intradiscal injection of 1 mL of 6.0 × 106 cells/disc combined with HA under real-time fluoroscopic guidance. Safety and feasibility were gauged using Visual Analogue Scale (VAS) pain and Oswestry Disability Index (ODI) scores and magnetic resonance imaging. All participants remained in the trial, with no reported adverse events linked to the procedure or stem cells. A successful outcome-marked by a minimum 2-point improvement in the VAS pain score and a 10-point improvement in ODI score from the start were observed in six participants. Although the modified Pfirrmann grade remained consistent across all participants, radiological improvements were evident in four patients. Specifically, two patients exhibited reduced high-intensity zones while another two demonstrated decreased disc protrusion. In conclusion, the intradiscal application of matrilin-3-primed ASC spheroids with HA is a safe and feasible treatment option for chronic discogenic LBP.
Collapse
Affiliation(s)
- Dong Hyun Lee
- Department of Neurosurgery, Spine Center, The Leon Wiltse Memorial Hospital, Suwon 16480, Republic of Korea;
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Kwang-Sook Park
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Hae Eun Shin
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Sung Bum Kim
- Department of Neurosurgery, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyejeong Choi
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Seong Bae An
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Joo Pyung Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea
| |
Collapse
|
12
|
Sargenti A, Pasqua S, Leu M, Dionisi L, Filardo G, Grigolo B, Gazzola D, Santi S, Cavallo C. Adipose Stromal Cell Spheroids for Cartilage Repair: A Promising Tool for Unveiling the Critical Maturation Point. Bioengineering (Basel) 2023; 10:1182. [PMID: 37892912 PMCID: PMC10603958 DOI: 10.3390/bioengineering10101182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Articular cartilage lacks intrinsic regenerative capabilities, and the current treatments fail to regenerate damaged tissue and lead only to temporary pain relief. These limitations have prompted the development of tissue engineering approaches, including 3D culture systems. Thanks to their regenerative properties and capacity to recapitulate embryonic processes, spheroids obtained from mesenchymal stromal cells are increasingly studied as building blocks to obtain functional tissues. The aim of this study was to investigate the capacity of adipose stromal cells to assemble in spheroids and differentiate toward chondrogenic lineage from the perspective of cartilage repair. Spheroids were generated by two different methods (3D chips vs. Ultra-Low Attachment plates), differentiated towards chondrogenic lineage, and their properties were investigated using molecular biology analyses, biophysical measurement of mass density, weight, and size of spheroids, and confocal imaging. Overall, spheroids showed the ability to differentiate by expressing specific cartilaginous markers that correlate with their mass density, defining a critical point at which they start to mature. Considering the spheroid generation method, this pilot study suggested that spheroids obtained with chips are a promising tool for the generation of cartilage organoids that could be used for preclinical/clinical approaches, including personalized therapy.
Collapse
Affiliation(s)
- Azzurra Sargenti
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Simone Pasqua
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Marco Leu
- abc biopply ag, 4500 Solothurn, Switzerland;
| | - Laura Dionisi
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Giuseppe Filardo
- Applied and Translational Research (ATR) Center, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Daniele Gazzola
- CellDynamics iSRL, 40136 Bologna, Italy; (A.S.); (S.P.); (L.D.); (D.G.)
| | - Spartaco Santi
- Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Unit of Bologna, CNR, 40136 Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Carola Cavallo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| |
Collapse
|
13
|
Peng Y, Jiang H, Zuo HD. Factors affecting osteogenesis and chondrogenic differentiation of mesenchymal stem cells in osteoarthritis. World J Stem Cells 2023; 15:548-560. [PMID: 37424946 PMCID: PMC10324504 DOI: 10.4252/wjsc.v15.i6.548] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/21/2023] [Accepted: 05/05/2023] [Indexed: 06/26/2023] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that often involves progressive cartilage degeneration and bone destruction of subchondral bone. At present, clinical treatment is mainly for pain relief, and there are no effective methods to delay the progression of the disease. When this disease progresses to the advanced stage, the only treatment option for most patients is total knee replacement surgery, which causes patients great pain and anxiety. As a type of stem cell, mesenchymal stem cells (MSCs) have multidirectional differentiation potential. The osteogenic differentiation and chondrogenic differentiation of MSCs can play vital roles in the treatment of OA, as they can relieve pain in patients and improve joint function. The differentiation direction of MSCs is accurately controlled by a variety of signaling pathways, so there are many factors that can affect the differentiation direction of MSCs by acting on these signaling pathways. When MSCs are applied to OA treatment, the microenvironment of the joints, injected drugs, scaffold materials, source of MSCs and other factors exert specific impacts on the differentiation direction of MSCs. This review aims to summarize the mechanisms by which these factors influence MSC differentiation to produce better curative effects when MSCs are applied clinically in the future.
Collapse
Affiliation(s)
- Yi Peng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hai Jiang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hou-Dong Zuo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Department of Radiology, Chengdu Xinhua Hospital, Chengdu 610067, Sichuan Province, China
| |
Collapse
|
14
|
Mélou C, Pellen-Mussi P, Novello S, Brézulier D, Novella A, Tricot S, Bellaud P, Chauvel-Lebret D. Spheroid Culture System, a Promising Method for Chondrogenic Differentiation of Dental Mesenchymal Stem Cells. Biomedicines 2023; 11:biomedicines11051314. [PMID: 37238984 DOI: 10.3390/biomedicines11051314] [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: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
The objective of the present work was to develop a three-dimensional culture model to evaluate, in a short period of time, cartilage tissue engineering protocols. The spheroids were compared with the gold standard pellet culture. The dental mesenchymal stem cell lines were from pulp and periodontal ligament. The evaluation used RT-qPCR and Alcian Blue staining of the cartilage matrix. This study showed that the spheroid model allowed for obtaining greater fluctuations of the chondrogenesis markers than for the pellet one. The two cell lines, although originating from the same organ, led to different biological responses. Finally, biological changes were detectable for short periods of time. In summary, this work demonstrated that the spheroid model is a valuable tool for studying chondrogenesis and the mechanisms of osteoarthritis, and evaluating cartilage tissue engineering protocols.
Collapse
Affiliation(s)
- Caroline Mélou
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| | - Pascal Pellen-Mussi
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Solen Novello
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| | - Damien Brézulier
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
| | - Agnès Novella
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Sylvie Tricot
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
| | - Pascale Bellaud
- CNRS, Inserm UMS Biosit, France BioImaging, Core Facility H2P2, University of Rennes, 35000 Rennes, France
| | - Dominique Chauvel-Lebret
- CNRS, ISCR (Institut des Sciences Chimiques de Rennes), University of Rennes, UMR 6226, 35000 Rennes, France
- Pôle d'Odontologie, Centre Hospitalier Universitaire de Rennes, 35033 Rennes, France
- UFR Odontologie, University of Rennes, 35043 Rennes, France
| |
Collapse
|
15
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
16
|
Sevastianov VI, Basok YB, Grigoriev AM, Nemets EA, Kirillova AD, Kirsanova LA, Lazhko AE, Subbot A, Kravchik MV, Khesuani YD, Koudan EV, Gautier SV. Decellularization of cartilage microparticles: Effects of temperature, supercritical carbon dioxide and ultrasound on biochemical, mechanical, and biological properties. J Biomed Mater Res A 2023; 111:543-555. [PMID: 36478378 DOI: 10.1002/jbm.a.37474] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/21/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
One of the approaches to restoring the structure of damaged cartilage tissue is an intra-articular injection of tissue-engineered medical products (TEMPs) consisting of biocompatible matrices loaded with cells. The most interesting are the absorbable matrices from decellularized tissues, provided that the cellular material is completely removed from them with the maximum possible preservation of the structure and composition of the natural extracellular matrix. The present study investigated the mechanical, biochemical, and biological properties of decellularized porcine cartilage microparticles (DCMps) obtained by techniques, differing only in physical treatments, such as freeze-thaw cycling (Protocol 1), supercritical carbon dioxide fluid (Protocol 2) and ultrasound (Protocol 3). Full tissue decellularization was achieved, as confirmed by the histological analysis and DNA quantification, though all the resultant DCMps had reduced glycosaminoglycans (GAGs) and collagen. The elastic modulus of all DCMp samples was also significantly reduced. Most notably, DCMps prepared with Protocol 3 significantly outperformed other samples in viability and the chondroinduction of the human adipose-derived stem cells (hADSCs), with a higher GAG production per DNA content. A positive ECM staining for type II collagen was also detected only in cartilage-like structures based on ultrasound-treated DCMps. The biocompatibility of a xenogenic DCMps obtained with Protocol 3 has been confirmed for a 6-month implantation in the thigh muscle tissue of mature rats (n = 18). Overall, the results showed that the porcine cartilage microparticles decellularized by a combination of detergents, ultrasound and DNase could be a promising source of scaffolds for TEMPs for cartilage reconstruction.
Collapse
Affiliation(s)
- Victor I Sevastianov
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia.,The Institute of Biomedical Research and Technology, Moscow, Russia
| | - Yulia B Basok
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Alexey M Grigoriev
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Evgeny A Nemets
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Alexandra D Kirillova
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Liudmila A Kirsanova
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia
| | - Aleksey E Lazhko
- Chemical Department, Lomonosov Moscow State University, Moscow, Russia
| | - Anastasia Subbot
- Laboratory of Fundamental Research in Ophtalmology, The Research Institute of Eye Diseases, Moscow, Russia
| | - Marina V Kravchik
- Laboratory of Fundamental Research in Ophtalmology, The Research Institute of Eye Diseases, Moscow, Russia
| | - Yusef D Khesuani
- Laboratory for Biotechnological Research "3D Bioprinting Solutions", Moscow, Russia
| | - Elizaveta V Koudan
- Center for Biomedical Engineering, National University of Science and Technology "MISIS", Moscow, Russia
| | - Sergey V Gautier
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, Moscow, Russia.,The Department of Transplantology and Artificial Organs, Faculty of Medicine, The Sechenov University, Moscow, Russia
| |
Collapse
|
17
|
Cryostructuring of Polymeric Systems: 63. † Synthesis of Two Chemically Tanned Gelatin-Based Cryostructurates and Evaluation of Their Potential as Scaffolds for Culturing of Mammalian Cells. Gels 2022; 8:gels8110695. [DOI: 10.3390/gels8110695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 11/17/2022] Open
Abstract
Various gelatin-containing gel materials are used as scaffolds for animal and human cell culturing within the fields of cell technologies and tissue engineering. Cryostructuring is a promising technique for the preparation of efficient macroporous scaffolds in biomedical applications. In the current study, two new gelatin-based cryostructurates were synthesized, their physicochemical properties and microstructure were evaluated, and their ability to serve as biocompatible scaffolds for mammalian cells culturing was tested. The preparation procedure included the dissolution of Type A gelatin in water, the addition of urea to inhibit self-gelation, the freezing of such a solution, ice sublimation in vacuo, and urea extraction with ethanol from the freeze-dried matter followed by its cross-linking in an ethanol medium with either carbodiimide or glyoxal. It was shown that in the former case, a denser cross-linked polymer phase was formed, while in the latter case, the macropores in the resultant biopolymer material were wider. The subsequent biotesting of these scaffolds demonstrated their biocompatibility for human mesenchymal stromal cells and HepG2 cells during subcutaneous implantation in rats. Albumin secretion and urea synthesis by HepG2 cells confirmed the possibility of using gelatin cryostructurates for liver tissue engineering.
Collapse
|
18
|
The Critical Role of Hypoxia in the Re-Differentiation of Human Articular Chondrocytes. Cells 2022; 11:cells11162553. [PMID: 36010629 PMCID: PMC9406483 DOI: 10.3390/cells11162553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/01/2022] [Accepted: 08/07/2022] [Indexed: 11/17/2022] Open
Abstract
The preservation of the chondrogenic phenotype and hypoxia-related physiological microenvironment are major challenges in the 2D culture of primary human chondrocytes. To address this problem, we develop a 3D culture system generating scaffold-free spheroids from human chondrocytes. Our results highlight the chondrogenic potential of cultured human articular chondrocytes in a 3D system combined with hypoxia independently of the cartilage source. After 14 days of culture, we developed spheroids with homogenous diameter and shape from hyaline cartilage donors. Spheroids generated in hypoxia showed a significantly increased glycosaminoglycans synthesis and up-regulated the expression of SOX9, ACAN, COL2A1, COMP, and SNAI1 compared to those obtained under normoxic conditions. Therefore, we conclude that spheroids developed under hypoxic conditions modulate the expression of chondrogenesis-related genes and native tissue features better than 2D cultures. Thus, this scaffold-free 3D culture system represents a novel in vitro model that can be used for cartilage biology research.
Collapse
|
19
|
Lee SY, Lee JW. 3D Spheroid Cultures of Stem Cells and Exosome Applications for Cartilage Repair. LIFE (BASEL, SWITZERLAND) 2022; 12:life12070939. [PMID: 35888029 PMCID: PMC9317836 DOI: 10.3390/life12070939] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022]
Abstract
Cartilage is a connective tissue that constitutes the structure of the body and consists of chondrocytes that produce considerable collagenous extracellular matrix and plentiful ground substances, such as proteoglycan and elastin fibers. Self-repair is difficult when the cartilage is damaged because of insufficient blood supply, low cellularity, and limited progenitor cell numbers. Therefore, three-dimensional (3D) culture systems, including pellet culture, hanging droplets, liquid overlays, self-injury, and spinner culture, have attracted attention. In particular, 3D spheroid culture strategies can enhance the yield of exosome production of mesenchymal stem cells (MSCs) when compared to two-dimensional culture, and can improve cellular restorative function by enhancing the paracrine effects of MSCs. Exosomes are membrane-bound extracellular vesicles, which are intercellular communication systems that carry RNAs and proteins. Information transfer affects the phenotype of recipient cells. MSC-derived exosomes can facilitate cartilage repair by promoting chondrogenic differentiation and proliferation. In this article, we reviewed recent major advances in the application of 3D culture techniques, cartilage regeneration with stem cells using 3D spheroid culture system, the effect of exosomes on chondrogenic differentiation, and chondrogenic-specific markers related to stem cell derived exosomes. Furthermore, the utilization of MSC-derived exosomes to enhance chondrogenic differentiation for osteoarthritis is discussed. If more mechanistic studies at the molecular level are conducted, MSC-spheroid-derived exosomes will supply a better therapeutic option to improve osteoarthritis.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea;
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Korea
- Correspondence: ; Tel.: +82-32-899-6516; Fax: +82-32-899-6039
| |
Collapse
|
20
|
The Induced Pluripotent Stem Cells in Articular Cartilage Regeneration and Disease Modelling: Are We Ready for Their Clinical Use? Cells 2022; 11:cells11030529. [PMID: 35159338 PMCID: PMC8834349 DOI: 10.3390/cells11030529] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/29/2022] [Accepted: 02/01/2022] [Indexed: 02/01/2023] Open
Abstract
The development of induced pluripotent stem cells has brought unlimited possibilities to the field of regenerative medicine. This could be ideal for treating osteoarthritis and other skeletal diseases, because the current procedures tend to be short-term solutions. The usage of induced pluripotent stem cells in the cell-based regeneration of cartilage damages could replace or improve on the current techniques. The patient’s specific non-invasive collection of tissue for reprogramming purposes could also create a platform for drug screening and disease modelling for an overview of distinct skeletal abnormalities. In this review, we seek to summarise the latest achievements in the chondrogenic differentiation of pluripotent stem cells for regenerative purposes and disease modelling.
Collapse
|
21
|
Shestovskaya MV, Bozhkova SA, Sopova JV, Khotin MG, Bozhokin MS. Methods of Modification of Mesenchymal Stem Cells and Conditions of Their Culturing for Hyaline Cartilage Tissue Engineering. Biomedicines 2021; 9:biomedicines9111666. [PMID: 34829895 PMCID: PMC8615732 DOI: 10.3390/biomedicines9111666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
The use of mesenchymal stromal cells (MSCs) for tissue engineering of hyaline cartilage is a topical area of regenerative medicine that has already entered clinical practice. The key stage of this procedure is to create conditions for chondrogenic differentiation of MSCs, increase the synthesis of hyaline cartilage extracellular matrix proteins by these cells and activate their proliferation. The first such works consisted in the indirect modification of cells, namely, in changing the conditions in which they are located, including microfracturing of the subchondral bone and the use of 3D biodegradable scaffolds. The most effective methods for modifying the cell culture of MSCs are protein and physical, which have already been partially introduced into clinical practice. Genetic methods for modifying MSCs, despite their effectiveness, have significant limitations. Techniques have not yet been developed that allow studying the effectiveness of their application even in limited groups of patients. The use of MSC modification methods allows precise regulation of cell culture proliferation, and in combination with the use of a 3D biodegradable scaffold, it allows obtaining a hyaline-like regenerate in the damaged area. This review is devoted to the consideration and comparison of various methods used to modify the cell culture of MSCs for their use in regenerative medicine of cartilage tissue.
Collapse
Affiliation(s)
- Maria V. Shestovskaya
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Svetlana A. Bozhkova
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
| | - Julia V. Sopova
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St. Petersburg, Russia
| | - Mikhail G. Khotin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
| | - Mikhail S. Bozhokin
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St. Petersburg, Russia; (M.V.S.); (J.V.S.); (M.G.K.)
- Vreden National Medical Research Center of Traumatology and Orthopedics, Academica Baykova Str., 8, 195427 St. Petersburg, Russia;
- Correspondence:
| |
Collapse
|
22
|
Sevastianov VI, Basok YB, Kirsanova LA, Grigoriev AM, Kirillova AD, Nemets EA, Subbot AM, Gautier SV. A Comparison of the Capacity of Mesenchymal Stromal Cells for Cartilage Regeneration Depending on Collagen-Based Injectable Biomimetic Scaffold Type. Life (Basel) 2021; 11:life11080756. [PMID: 34440500 PMCID: PMC8400656 DOI: 10.3390/life11080756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/21/2021] [Accepted: 07/25/2021] [Indexed: 01/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have shown a high potential for cartilage repair. Collagen-based scaffolds are used to deliver and retain cells at the site of cartilage damage. The aim of the work was a comparative analysis of the capacity of the MSCs from human adipose tissue to differentiate into chondrocytes in vitro and to stimulate the regeneration of articular cartilage in an experimental model of rabbit knee osteoarthrosis when cultured on microheterogenic collagen-based hydrogel (MCH) and the microparticles of decellularized porcine articular cartilage (DPC). The morphology of samples was evaluated using scanning electron microscopy and histological staining methods. On the surface of the DPC, the cells were distributed more uniformly than on the MCH surface. On day 28, the cells cultured on the DPC produced glycosaminoglycans more intensely compared to the MCH with the synthesis of collagen type II. However, in the experimental model of osteoarthrosis, the stimulation of the cartilage regeneration was more effective when the MSCs were administered to the MCH carrier. The present study demonstrates the way to regulate the action of the MSCs in the area of cartilage regeneration: the MCH is more conducive to stimulating cartilage repair by the MSCs, while the DPC is an inducer for a formation of a cartilage-like tissue by the MSCs in vitro.
Collapse
Affiliation(s)
- Victor I. Sevastianov
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
- Correspondence: ; Tel.: +7-916-173-41-84
| | - Yulia B. Basok
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
| | - Ludmila A. Kirsanova
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
| | - Alexey M. Grigoriev
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
| | - Alexandra D. Kirillova
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
| | - Evgeniy A. Nemets
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
| | - Anastasia M. Subbot
- Laboratory of Fundamental Research in Ophtalmology, The Research Institute of Eye Diseases, 11A, B Rossolimo St., 119021 Moscow, Russia;
| | - Sergey V. Gautier
- Department for Biomedical Technologies and Tissue Engineering, The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 1 Shchukinskaja St., 123182 Moscow, Russia; (Y.B.B.); (L.A.K.); (A.M.G.); (A.D.K.); (E.A.N.); (S.V.G.)
- Department of Transplantology and Artificial Organs, Faculty of Medicine, The Sechenov University, 8-2 Trubetskaya St., 119991 Moscow, Russia
| |
Collapse
|