1
|
Sutthiwanjampa C, Kang SH, Kim MK, Hwa Choi J, Kim HK, Woo SH, Bae TH, Kim WJ, Kang SH, Park H. Tumor necrosis factor-α-treated human adipose-derived stem cells enhance inherent radiation tolerance and alleviate in vivo radiation-induced capsular contracture. J Adv Res 2025; 72:433-449. [PMID: 39019109 DOI: 10.1016/j.jare.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024] Open
Abstract
INTRODUCTION Post-mastectomy radiotherapy plays a crucial role in breast cancer treatment but can lead to an inflammatory response causing soft tissue damage, particularly radiation-induced capsular contracture (RICC), impacting breast reconstruction outcomes. Adipose-derived stem cells (ADSCs), known for their regenerative potential via paracrine capacity, exhibit inherent radiotolerance. The influence of tumor necrosis factor-alpha (TNF-α) on ADSCs has been reported to enhance the paracrine effect of ADSCs, promoting wound healing by modulating inflammatory responses. OBJECTIVE This study investigates the potential of TNF-α-treated human ADSCs (T-hASCs) on silicone implants to alleviate RICC, hypothesizing to enhance suppressive effects on RICC by modulating inflammatory responses in a radiation-exposed environment. METHODS In vitro, T-hASCs were cultured on various surfaces to assess viability after exposure to radiation up to 20 Gy. In vivo, T-hASC and non-TNF-α-treated hASC (C-hASCs)-coated membranes were implanted in mice before radiation exposure, and an evaluation of the RICC mitigation took place 4 and 8 weeks after implantation. In addition, the growth factors released from T-hASCs were assessed. RESULTS In vitro, hASCs displayed significant radiotolerance, maintaining consistent viability after exposure to 10 Gy. TNF-α treatment further enhanced radiation tolerance, as evidenced by significantly higher viability than C-hASCs at 20 Gy. In vivo, T-hASC-coated implants effectively suppressed RICC, reducing capsule thickness. T-hASCs exhibited remarkable modulation of the inflammatory response, suppressing M1 macrophage polarization while enhancing M2 polarization. The elevated secretion of vascular endothelial growth factor from T-hASCs is believed to induce macrophage polarization, potentially reducing RICC. CONCLUSION This study establishes T-hASCs as a promising strategy for ameliorating the adverse effects experienced by breast reconstruction patients after mastectomy and radiation therapy. The observed radiotolerance, anti-fibrotic effects, and immune modulation suggest the possibility of enhancing patient outcomes and quality of life. Further research and clinical trials are warranted for broader clinical uses.
Collapse
Affiliation(s)
- Chanutchamon Sutthiwanjampa
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Seung Hyun Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Mi Kyung Kim
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Departments of Pathology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Jin Hwa Choi
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Radiation Oncology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Han Koo Kim
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Soo Hyun Woo
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Tae Hui Bae
- Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea
| | - Woo Joo Kim
- Department of Plastic Surgery, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-si, Gyeonggi-do 14353, Republic of Korea
| | - Shin Hyuk Kang
- College of Medicine, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea; Department of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, 102 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06973, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, 84 Heukseok-ro, Heukseok-dong, Dongjak-gu, Seoul 06974, Republic of Korea.
| |
Collapse
|
2
|
Schneider RS, Nieves EB, Aggarwal B, Bowles-Welch AC, Stevens HY, Kippner LE, Boden SD, Mautner K, Drissi H, Roy K, Lam WA, Sinha S, García AJ. On-chip 3D potency assay for prediction of clinical outcomes for cell therapy candidates for osteoarthritis. Nat Commun 2025; 16:4915. [PMID: 40425577 DOI: 10.1038/s41467-025-60158-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
The lack of clinically predictive potency assays for cell products significantly impedes translation of these therapies. Here, we describe a microfluidic on-chip 3D system for rapid evaluation of a subset of patient-derived bone marrow aspirate concentrate (BMAC) samples used in a phase 3 multicenter trial (NCT03818737) evaluating autologous cells for relieving knee osteoarthritis pain. BMAC clinical samples cultured in the on-chip 3D system exhibit elevated levels of immunomodulatory and trophic proteins compared to 2D culture. Using analyte information from in vitro assays and patient-matched clinical data, we build linear regression prediction models for clinical outcomes. We demonstrate improved clinical prediction by cross-validation accuracy for the on-chip 3D platform compared to 2D culture. Additionally, on-chip 3D assay metrics display higher correlative power with patient pain scores compared to the 2D assay. This study establishes a potency assay with improved prediction power to accelerate translation of cell therapies.
Collapse
Affiliation(s)
- Rebecca S Schneider
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elisa B Nieves
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Bhavay Aggarwal
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Annie C Bowles-Welch
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hazel Y Stevens
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA
| | - Linda E Kippner
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, USA
| | - Scott D Boden
- Department of Orthopaedics, Emory University, Atlanta, GA, USA
| | - Kenneth Mautner
- Department of Orthopaedics, Emory University, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University, Atlanta, GA, USA
| | - Krishnendu Roy
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Wilbur A Lam
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Saurabh Sinha
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Parsons AM, Ahsan N, Darling EM. Identifying Immunomodulatory Subpopulations of Adipose Stromal Vascular Fraction and Stem/Stromal Cells Through Single-Cell Transcriptomics and Bulk Proteomics. Stem Cell Rev Rep 2025:10.1007/s12015-025-10889-6. [PMID: 40366552 DOI: 10.1007/s12015-025-10889-6] [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] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
A primary therapeutic characteristic of mesenchymal stem/stromal cells (MSCs) is their immunomodulatory activity. Adipose-derived stem/stromal cells (ASCs) are an abundant and easily isolated source of MSCs shown to have high immunosuppressive activity, making them attractive for therapy. Understanding the heterogeneous immunomodulatory potential of ASCs within the stromal vascular fraction (SVF) of adipose tissue could better inform treatment strategies. In this study, we integrate single-cell RNA sequencing (scRNA seq) with bulk proteomics to characterize subpopulations of SVF-derived ASCs that are phenotypically similar to cytokine-licensed, cultured ASCs. To better define the licensing process, we present scRNA seq and bulk proteomics data of cultured (P2) ASCs exposed to inflammatory cytokines, showing enrichment of pathways related to inflammation and apoptosis that positively correlate to the cytokine-mediated, trajectory-derived pseudotime. Using the Scissor algorithm, we integrate the proteomics data with uncultured (P0) SVF scRNA seq data, identifying an ASC subpopulation that is phenotypically like the cytokine-stimulated ASCs (Scissor-positive). Interactome analysis identifies Scissor-positive ASCs as stress adaptive immune regulators that function through IL6 and broad SEMA4 interactions and higher Visfatin signaling, while Scissor-negative ASCs show strong signatures of ECM remodeling through FN1 and immunosuppression through THY1 and MIF signaling. Our multimodal, integrative approach enabled identification of previously unidentified, distinct ASC subpopulations with differing immunomodulatory phenotypes that are present in, and can potentially be selected from, P0 SVF ASCs.
Collapse
Affiliation(s)
- Adrienne M Parsons
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 02115, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, 02115, MA, USA
| | - Nagib Ahsan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73104, USA
- Mass Spectrometry, Proteomics and Metabolomics Core Facility, University of Oklahoma, Norman, OK, 73104, USA
| | - Eric M Darling
- Department of Pathology and Laboratory Medicine, Brown University, 171 Meeting St, Providence, RI, 02912, USA.
- School of Engineering, Brown University, Providence, RI, 02912, USA.
- Department of Orthopaedics, Brown University, Brown University, RI, 02912, USA.
- Institute for Biology, Engineering, and Medicine, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
4
|
Bukauskas A, Jucaitienė R, Stoškus M, Valčeckienė V, Bušmaitė G, Slobinas A, Davainis L, Šlepikienė I, Trociukas I, Pečeliūnas V, Griškevičius L, Žučenka A. Mesenchymal stromal cells for steroid-refractory biopsy-proven grade III-IV acute Graft-versus-Host Disease with predominant gastrointestinal involvement. Front Immunol 2025; 16:1600019. [PMID: 40433379 PMCID: PMC12106019 DOI: 10.3389/fimmu.2025.1600019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction Steroid-refractory acute Graft-versus-Host Disease (SR-aGVHD) is a potentially fatal complication occurring in approximately 60-70% of severe grade III-IV GVHD cases, with a higher incidence in patients with gastrointestinal (GI) involvement. GI aGVHD is associated with poor prognosis, with a 2-year overall survival (OS) rate of only 25% in patients with stage 3-4 GI involvement. Mesenchymal stromal cells (MSC) have emerged as a promising therapeutic option due to their favorable efficacy and safety profile. However, data on bone marrow (BM)-derived MSC use in biopsy-proven grade III-IV SR-aGVHD with GI involvement, particularly in stage 3-4 cases, remain limited. Methods This prospective, observational, single-arm, single-center study assessed the efficacy and safety of BM-derived MSC for treating adult patients with biopsy-proven grade III-IV SR-aGVHD with predominant GI involvement. Early (1st-2nd) passage BM-derived MSC were administered weekly at a target dose of 1x106 MSC/kg in two regimens: up to three (MSC3) and six doses (MSC6). Results Fifty-seven adult patients with biopsy-proven III-IV grade SR-aGVHD (93% with GI involvement) received MSC treatment. The overall response rate (ORR) was 39% and 42% on Days 14 and 28, respectively, with no significant differences between the two MSC groups (Day 28 ORR 38% for MSC3 and 44% for MSC6). In patients with stage 3-4 GI involvement, the ORR was 26% and 36% at the corresponding time points with comparable efficacy between the two MSC groups (Day 28 ORR 31% for MSC3 and 38% for MSC6). Day 14 and Day 28 responders had significantly higher OS compared to non-responders (52% vs. 7%, p=0.000; 54% vs. 5%, p=0.000), with a comparable OS benefit observed in patients with stage 3-4 GI involvement (45% vs. 8%, p=0.005; 42% vs. 6%, p=0.005), respectively. MSC treatment had a favorable safety profile. The one, 5 and 10-year OS rates were 27%, 24%, and 24%, respectively. Conclusions The grade III-IV SR-aGVHD patients, including cases with biopsy-proven severe GI involvement, had significantly better clinical outcomes if responses to MSC treatment were observed on Days 14 and 28. Intensified MSC administration schedule has failed to improve the clinical outcomes. MSC studies focusing on aGVHD prevention and (or) first-line treatment in combination with other agents should be considered.
Collapse
Affiliation(s)
- Adomas Bukauskas
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Renata Jucaitienė
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Mindaugas Stoškus
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Vilma Valčeckienė
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Greta Bušmaitė
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Artūras Slobinas
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Hematology and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Linas Davainis
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Inga Šlepikienė
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Igoris Trociukas
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Valdas Pečeliūnas
- Department of Hematology and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Laimonas Griškevičius
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Hematology and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Andrius Žučenka
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Department of Hematology and Oncology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
5
|
Faircloth TU, Temple S, Parr R, Soma A, Massoumi H, Jalilian E, Djalilian AR, Hematti P, Rajan D, Chinnadurai R. Human cornea-derived mesenchymal stromal cells inhibit T cells through indoleamine 2,3 dioxygenase. Cytotherapy 2025; 27:597-608. [PMID: 39891632 PMCID: PMC12097958 DOI: 10.1016/j.jcyt.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Defining the mechanism of immune modulation by mesenchymal stromal cells (MSCs) from distinct anatomical tissues is of great translational interest. The human cornea is an immunologically privileged organ, and the mechanism of immunoregulation of cornea-derived MSCs (cMSCs) is currently unknown. We investigated cMSCs derived from the corneas of 5 independent human donorS for their fitness and mechanism of action in suppressing T cells. cMSCs display the immunophenotype CD45-CD73+CD105+CD90+CD44+ and robust in vitro growth. 30-plex secretome analysis identified that cMSCs innately secrete specific molecules in a dose-dependent manner. cMSCs do not express or upregulate costimulatory but do upregulate coinhibitory molecules upon stimulation with interferon γ (IFNγ). cMSCs inhibit T-cell proliferation in contact-dependent co-cultures, which can be predicted by a unique secretome signature. In addition, co-culturing in a 2-chamber transwell system has demonstrated that cMSCs also inhibit T-cell proliferation in a non-contact-dependent manner. Mechanistic analysis has demonstrated that activated T cells effectively induce indoleamine 2,3-dioxygenase (IDO) but not other enzymes of the tryptophan metabolic pathway in cMSCs. Silencing of IDO in cMSCs reduces their fitness to suppress T cells. These results provide evidence that in cMSCs, one of the principal mechanisms of immunosuppression on T cells is through IDO. These results suggest that MSCs derived from the human cornea display immunoregulatory properties and, thus, may play a role in maintaining the immune-privileged niche of the cornea.
Collapse
Affiliation(s)
- Tyler U Faircloth
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA
| | - Sara Temple
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA
| | - Rhett Parr
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA
| | - Alyssa Soma
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA
| | - Hamed Massoumi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Elmira Jalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Peiman Hematti
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Devi Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, Georgia, USA.
| |
Collapse
|
6
|
Hung CY, Hsueh TY, Rethi L, Lu HT, Chuang AEY. Advancements in regenerative medicine: a comprehensive review of stem cell and growth factor therapies for osteoarthritis. J Mater Chem B 2025; 13:4494-4526. [PMID: 40042377 DOI: 10.1039/d4tb01769b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Osteoarthritis (OA) is a widely encountered degenerative joint disorder marked by gradual cartilage deterioration, inflammation, and pain, which collectively impose considerable strain on global healthcare systems. While traditional therapies typically offer relief from symptoms, they do not tackle the core pathophysiological aspects of the disease. Regenerative medicine has recently risen as a promising field for addressing OA, capitalizing on the regenerative capabilities of stem cells and growth factors to foster tissue healing and renewal. This thorough review delves into the most recent progress in stem cell and growth factor treatments for OA, covering preclinical studies, clinical trials, and novel technological developments. We discuss the diverse origins of stem cells, such as mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and adipose-derived stem cells (ASCs), underscoring their therapeutic actions and effectiveness in both preclinical and clinical environments. Moreover, we explore contributions of growth factors like transforming growth factor (TGF)-β, platelet-derived growth factor (PDGF), and insulin-like growth factor (IGF) in modifying OA's pathology and enhancing tissue restoration. Additionally, this review discusses the hurdles and constraints tied to current regenerative strategies, including the standardization of cell sources, the refinement of delivery techniques, and considerations for long-term safety. By meticulously assessing the latest research outcomes and technological breakthroughs, this review aims to shed light on the potential of stem cell and growth factor therapies as forthcoming therapeutic options for OA, thereby propelling forward the domain of regenerative medicine and enhancing clinical results for individuals afflicted with this incapacitating ailment.
Collapse
Affiliation(s)
- Chen-Yuan Hung
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tai-Yuan Hsueh
- School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Lekshmi Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
| | - Hsien-Tsung Lu
- Department of Orthopedics, Taipei Medical University Hospital, Taipei City 11031, Taiwan
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- International PhD Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Andrew E-Y Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, Taiwan
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, Taiwan
| |
Collapse
|
7
|
Yanuar A, Agustina H, Antarianto RD, Hidajat NN, Mahyuddin AI, Dilogo IH, Budhiparama NC, Atik N. Extracellular Vesicles from Adipose-Derived Mesenchymal Stem Cells Improve Ligament-Bone Integration After Anterior Cruciate Ligament Primary Repair in Rabbit. Biomolecules 2025; 15:396. [PMID: 40149932 PMCID: PMC11940348 DOI: 10.3390/biom15030396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUNDS In this research, we want to find out whether extracellular vesicles (EVs) from adipose-derived mesenchymal stem cells (MSCs) can improve ligament-bone integration after primary Anterior Cruciate Ligament (ACL) repair by performing immunological and biomechanical tests. METHODS All of the rabbits underwent ACL resection at the proximal attachment to the femur bone, and then were divided into four groups. We performed an ELISA examination from the tissue at the bone-ligament interface of iNOS, CD206, MMP-3, and TIMP-1 to evaluate their levels at the inflammatory stage at the end of the first week. Immunoexpression of type I and III collagen and failure load biomechanical tests were performed at the end of the sixth week. RESULT The group that underwent ACL repair with EVs augmentation had significantly higher levels of CD206, significantly lower MMP-3 levels, and significantly higher TIMP-1 levels in the first week. The iNOS levels in the group that underwent ACL repair with EVs augmentation were significantly different compared to the control group that did not receive any. The number of type I collagen fibers and the failure load levels in the group that underwent ACL repair with EVs augmentation were significantly higher. CONCLUSIONS EVs from adipose-derived MSCs can improve the outcome of primary ACL repair in rabbits by regulating the inflammatory process during the healing period.
Collapse
Affiliation(s)
- Andre Yanuar
- Doctoral Program, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia;
- Department of Orthopaedic and Traumatology, Santo Borromeus Hospital, Bandung 40132, Indonesia
| | - Hasrayati Agustina
- Department of Pathology Anatomy, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia;
| | - Radiana Dhewayani Antarianto
- Stem Cell and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia; (R.D.A.); (I.H.D.)
- Department of Histology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| | - Nucki Nursjamsi Hidajat
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia;
| | - Andi Isra Mahyuddin
- Faculty of Mechanical and Aerospace Engineering, Institut Teknologi Bandung (ITB), Bandung 40132, Indonesia;
| | - Ismail Hadisoebroto Dilogo
- Stem Cell and Tissue Engineering Research Cluster, Indonesian Medical Education and Research Institute, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia; (R.D.A.); (I.H.D.)
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universitas Indonesia/Cipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia
- Stem Cell Integrated Medical Technology Service Unit, Faculty of Medicine, Universitas Indonesia/ipto Mangunkusumo General Hospital, Jakarta 10430, Indonesia
| | - Nicolaas Cyrillus Budhiparama
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia
- Department of Orthopaedics, Leiden University Medical Centre, 2333 ZA Leiden, The Netherlands
| | - Nur Atik
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
| |
Collapse
|
8
|
Bukulmez H, Dennis AT, Reese-Koc J, Sieg SF, Clagett B, Kleinsorge-Block S, Somoza-Palacios R, Singer N, Chance M, Highland KB, Emancipator SN. Trained mesenchymal stromal cell-based therapy HXB-319 for treating diffuse alveolar hemorrhage in a pristane-induced murine model. Stem Cells 2025; 43:sxae078. [PMID: 39560076 PMCID: PMC11878545 DOI: 10.1093/stmcls/sxae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024]
Abstract
INTRODUCTION Mesenchymal stromal cells (MSCs) can modulate immune responses and suppress inflammation in autoimmune diseases. Although their safety has been established in clinical trials, the efficacy of MSCs is inconsistent due to variability in potency among different preparations and limited specificity in targeting mechanisms driving autoimmune diseases. METHODS We utilized high-dimensional design of experiments methodology to identify factor combinations that modulate gene expression by MSCs to mitigate inflammation. This led to a novel MSC-based cell therapy, HXB-319. Its anti-inflammatory properties were validated in vitro by flow cytometry, RT-PCR, and mass spectrophotometry. To evaluate in vivo efficacy, we treated a diffuse alveolar hemorrhage (DAH) mouse model (C57Bl/6). Seven days post-DAH induction with pristane, mice received either MSCs or HXB-319 (2X106 cells, IP). On day 14, peritoneal lavage fluid (PLF) and lung tissue were collected for flow cytometry, histopathological examination, and mRNA. RESULTS HXB-319 increased gene expression levels of anti-inflammatory, angiogenic, and anti-fibrotic factors (eg, TSG-6, VEGF, and HGF). KEGG pathway analysis confirmed significant activation of relevant anti-inflammatory, angiogenic, and anti-fibrotic proteins, corroborating RT-PCR results. In the DAH model, HXB-319 significantly reduced lung inflammation and alveolar hemorrhage compared to MSC-treated and untreated DAH mice. HXB-319 treatment also significantly decreased neutrophils, plasmacytoid dendritic cells, and RORγT cells, increased FoxP3+ cells in PLF, and reversed alterations in mRNA encoding IL-6, IL-10, and TSG-6 in lung tissue compared to DAH mice. CONCLUSION HXB-319 effectively controls inflammation and prevents tissue damage in pristine-induced DAH, highlighting its therapeutic potential for autoimmune inflammatory diseases.
Collapse
Affiliation(s)
- Hulya Bukulmez
- Division of Pediatric Rheumatology, Department of Pediatrics, Metrohealth Medical Center, Case Western Reserve University, Cleveland, OH, United States
| | - Adrienne T Dennis
- Division of Pediatric Rheumatology, Department of Pediatrics, Metrohealth Medical Center, Case Western Reserve University, Cleveland, OH, United States
| | - Jane Reese-Koc
- Cellular Therapy Operations and Quality, National Center for Regenerative Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Scott F Sieg
- Immunology Flow Core, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Brian Clagett
- Immunology Flow Core, Department of Medicine, Division of Infectious Diseases and HIV Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sarah Kleinsorge-Block
- Cellular Therapy Operations and Quality, National Center for Regenerative Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Rodrigo Somoza-Palacios
- Skeletal Research Center, Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Nora Singer
- Division of Rheumatology, Department of Internal Medicine, Metrohealth Medical Center, Case Western Reserve University, Cleveland, OH, United States
| | - Mark Chance
- Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Kristin B Highland
- Department of Pulmonary/Critical Care, Rheumatic Lung Disease Program, Cleveland Clinic Foundation, Case Western Reserve University, Cleveland, OH, United States
| | - Steven N Emancipator
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
9
|
Hodgson-Garms M, Moore MJ, Martino MM, Kelly K, Frith JE. Proteomic profiling of iPSC and tissue-derived MSC secretomes reveal a global signature of inflammatory licensing. NPJ Regen Med 2025; 10:7. [PMID: 39905050 PMCID: PMC11794695 DOI: 10.1038/s41536-024-00382-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 12/03/2024] [Indexed: 02/06/2025] Open
Abstract
Much of the therapeutic potential of mesenchymal stromal cells (MSCs) is underpinned by their secretome which varies significantly with source, donor and microenvironmental cues. Understanding these differences is essential to define the mechanisms of MSC-based tissue repair and optimise cell therapies. This study analysed the secretomes of bone-marrow (BM.MSCs), umbilical-cord (UC.MSCs), adipose-tissue (AT.MSCs) and clinical/commercial-grade induced pluripotent stem cell-derived MSCs (iMSCs), under resting and inflammatory licenced conditions. iMSCs recapitulated the inflammatory licensing process, validating their comparability to tissue-derived MSCs. Overall, resting secretomes were defined by extracellular matrix (ECM) and pro-regenerative proteins, while licensed secretomes were enriched in chemotactic and immunomodulatory proteins. iMSC and UC.MSC secretomes contained proteins indicating proliferative potential and telomere maintenance, whereas adult tissue-derived secretomes contained fibrotic and ECM-related proteins. The data and findings from this study will inform the optimum MSC source for particular applications and underpin further development of MSC therapies.
Collapse
Affiliation(s)
- Margeaux Hodgson-Garms
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia.
- Cynata Therapeutics, Melbourne, VIC, Australia.
| | - Matthew J Moore
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia
| | - Mikaël M Martino
- Australian Regenerative Medicine Institute, Melbourne, VIC, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | | | - Jessica E Frith
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia.
- Australian Regenerative Medicine Institute, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Pierro M, Thébaud B. Cell-based strategies for the treatment of injury to the developing lung. THE LUNG 2025:403-426. [DOI: 10.1016/b978-0-323-91824-4.00020-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Wang J, Zhou Y, Donohoe E, Canning A, Moosavizadeh S, Ryan AE, Ritter T. Immunomodulatory potential of cytokine-licensed human bone marrow-derived mesenchymal stromal cells correlates with potency marker expression profile. Stem Cells 2024; 42:1040-1054. [PMID: 39208292 PMCID: PMC11630899 DOI: 10.1093/stmcls/sxae053] [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: 03/12/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024]
Abstract
Cytokine(s) pre-activation/licensing is an effective way to enhance the immunomodulatory potency of mesenchymal stromal cells (MSCs). Currently, IFN-γ licensing received the most attention in comparison with other cytokines. After licensing human bone marrow-derived MSCs with pro-/anti-inflammatory cytokines IFN-γ, IL-1β, TNF-α, TGF-β1 alone or in combination, the in vitro immunomodulatory potency of these MSCs was studied by incubating with allogeneic T cells and macrophage-like THP-1 cells. In addition, immunomodulation-related molecules filtered by bioinformatics, complement 1 subcomponent (C1s), and interferon-induced GTP-binding protein Mx2 (MX2), were studied to verify whether to reflect the immunomodulatory potency. Herein, we reported that different cytokines cause different effects on the function of MSC. While TGF-β1 licensing enhances the capacity of MSCs to induce T cells with an immunosuppressive phenotype, IFN-γ-licensing strengthens the inhibitory effect of MSC on T cell proliferation. Both TGF-β1 and IFN-γ licensing can enhance the effect of MSC on reducing the expression of pro-inflammatory cytokines by M1 macrophage-like THP-1 cells. Interestingly, IFN-γ upregulates potential potency markers extracellular C1s and kynurenine (KYN) and intracellular MX2. These 3 molecules have the potential to reflect mesenchymal stromal cell immunomodulatory potency. In addition, we reported that there is a synergistic effect of TGF-β1 and IFN-γ in immunomodulation.
Collapse
Affiliation(s)
- Jiemin Wang
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
| | - Yingying Zhou
- Changsha Centre for Disease Control and Prevention, Changsha, Hunan Province 410011, People’s Republic of China
| | - Ellen Donohoe
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
| | - Aoife Canning
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
| | - Seyedmohammad Moosavizadeh
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
- CURAM Centre for Research in Medical Devices, University of Galway, Galway H91FD82, Ireland
| | - Aideen E Ryan
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
- CURAM Centre for Research in Medical Devices, University of Galway, Galway H91FD82, Ireland
- Discipline of Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway H91TK33, Ireland
| | - Thomas Ritter
- Regenerative Medicine Institute, School of Medicine, University of Galway, Galway H91FD82, Ireland
- CURAM Centre for Research in Medical Devices, University of Galway, Galway H91FD82, Ireland
| |
Collapse
|
12
|
AlOraibi S, Taurin S, Alshammary S. Advancements in Umbilical Cord Biobanking: A Comprehensive Review of Current Trends and Future Prospects. Stem Cells Cloning 2024; 17:41-58. [PMID: 39655226 PMCID: PMC11626973 DOI: 10.2147/sccaa.s481072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Biobanking has emerged as a transformative concept in advancing the medical field, particularly with the exponential growth of umbilical cord (UC) biobanking in recent decades. UC blood and tissue provide a rich source of primitive hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs) for clinical transplantation, offering distinct advantages over alternative adult stem cell sources. However, to fully realize the therapeutic potential of UC-derived stem cells and establish a comprehensive global UC-biobanking network, it is imperative to optimize and standardize UC processing, cryopreservation methods, quality control protocols, and regulatory frameworks, alongside developing effective consent provisions. This review aims to comprehensively explore recent advancements in UC biobanking, focusing on the establishment of rigorous safety and quality control procedures, the standardization of biobanking operations, and the optimization and automation of UC processing and cryopreservation techniques. Additionally, the review examines the expanded clinical applications of UC stem cells, addresses the challenges associated with umbilical cord biobanking and UC-derived stem cell therapies, and discusses the promising role of artificial intelligence (AI) in enhancing various operational aspects of biobanking, streamlining data processing, and improving data analysis accuracy while ensuring compliance with safety and quality standards. By addressing these critical areas, this review seeks to provide insights into the future direction of UC biobanking and its potential to significantly impact regenerative medicine.
Collapse
Affiliation(s)
- Sahar AlOraibi
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Sebastien Taurin
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| | - Sfoug Alshammary
- Molecular Medicine Department, Princess Al Jawhara Center for Molecular Medicine, Genetics, and Hereditary Diseases, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
13
|
Gao P, Kajiya M, Motoike S, Ikeya M, Yang J. Application of mesenchymal stem/stromal cells in periodontal regeneration: Opportunities and challenges. JAPANESE DENTAL SCIENCE REVIEW 2024; 60:95-108. [PMID: 38314143 PMCID: PMC10837070 DOI: 10.1016/j.jdsr.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/06/2023] [Accepted: 01/15/2024] [Indexed: 02/06/2024] Open
Abstract
Guided tissue regeneration (GTR) has been widely used in the periodontal treatment of intrabony and furcation defects for nearly four decades. The treatment outcomes have shown effectiveness in reducing pocket depth, improving attachment gain and bone filling in periodontal tissue. Although applying GTR could reconstruct the periodontal tissue, the surgical indications are relatively narrow, and some complications and race ethic problems bring new challenges. Therefore, it is challenging to achieve a consensus concerning the clinical benefits of GTR. With the appearance of stem cell-based regenerative medicine, mesenchymal stem/stromal cells (MSCs) have been considered a promising cell resource for periodontal regeneration. In this review, we highlight preclinical and clinical periodontal regeneration using MSCs derived from distinct origins, including non-odontogenic and odontogenic tissues and induced pluripotent stem cells, and discuss the transplantation procedures, therapeutic mechanisms, and concerns to evaluate the effectiveness of MSCs.
Collapse
Affiliation(s)
- Pan Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of General Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Mikihito Kajiya
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Souta Motoike
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Makoto Ikeya
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Jingmei Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
14
|
Wang X, Wang Q, Xia Z, Yang Y, Dai X, Zhang C, Wang J, Xu Y. Mesenchymal stromal cell therapies for traumatic neurological injuries. J Transl Med 2024; 22:1055. [PMID: 39578845 PMCID: PMC11583761 DOI: 10.1186/s12967-024-05725-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/01/2024] [Indexed: 11/24/2024] Open
Abstract
Improved treatment options are urgently needed for neurological injuries resulting from trauma or iatrogenic events causing long-term disabilities that severely impact patients' quality of life. In vitro and animal studies have provided promising proof-of-concept examples of regenerative therapies using mesenchymal stromal cells (MSC) for a wide range of pathological conditions. Over the previous decade, various MSC-based therapies have been investigated in clinical trials to treat traumatic neurological injuries. However, while the safety and feasibility of MSC treatments has been established, the patient outcomes in these studies have not demonstrated significant success in the translation of MSC regenerative therapy for the treatment of human brain and spinal cord injuries. Herein, we have reviewed the literature and ongoing registered trials on the application of MSC for the treatment of traumatic brain injury, traumatic spinal cord injury, and peripheral nerve injury. We have focused on the shortcomings and technological hurdles that must be overcome to further advance clinical research to phase 3 trials, and we discuss recent advancements that represent potential solutions to these obstacles to progress.
Collapse
Affiliation(s)
- Xiujuan Wang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Qian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China
| | - Ziyao Xia
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Ying Yang
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China
| | - Xunan Dai
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| | - Jiaxian Wang
- HELP Therapeutics Co., Ltd, No. 568 Longmian Avenue, Jiangning District, Nanjing, 211166, Jiangsu Province, China.
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, E12 Avenida da Universidade, Macau, 519000, SAR, China.
| | - Yongsheng Xu
- Technology Department, Tianjin Everunion Biotechnology Co., Ltd, SOHO Nexus Center, No. 19A East 3rd Ring North Road, Chaoyang District, Beijing, 100020, China.
- Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, No. 49 North Garden Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
15
|
Fang WH, Vangsness CT. Orthobiologic Products: Preservation Options for Orthopedic Research and Clinical Applications. J Clin Med 2024; 13:6577. [PMID: 39518716 PMCID: PMC11546119 DOI: 10.3390/jcm13216577] [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: 09/11/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The biological products used in orthopedics include musculoskeletal allografts-such as bones, tendons, ligaments, and cartilage-as well as biological therapies. Musculoskeletal allografts support the body's healing process by utilizing preserved and sterilized donor tissue. These allografts are becoming increasingly common in surgical practice, allowing patients to avoid more invasive procedures and the risks associated with donor site morbidity. Bone grafting is one of the most frequently used procedures in orthopedics and traumatology. Biologic approaches aim to improve clinical outcomes by enhancing the body's natural healing capacity and reducing inflammation. They serve as an alternative to surgical interventions. While preliminary results from animal studies and small-scale clinical trials have been promising, the field of biologics still lacks robust clinical evidence supporting their efficacy. Biological therapies include PRP (platelet-rich plasma), mesenchymal stem cells (MSCs)/stromal cells/progenitor cells, bone marrow stem/stromal cells (BMSCs), adipose stem/stromal cells/progenitor cells (ASCs), cord blood (CB), and extracellular vesicles (EVs), including exosomes. The proper preservation and storage of these cellular therapies are essential for future use. Preservation techniques include cryopreservation, vitrification, lyophilization, and the use of cryoprotective agents (CPAs). The most commonly used CPA is DMSO (dimethyl sulfoxide). The highest success rates and post-thaw viability have been achieved by preserving PRP with a rate-controlled freezer using 6% DMSO and storing other cellular treatments using a rate-controlled freezer with 5% or 10% DMSO as the CPA. Extracellular vesicles (EVs) have shown the best results when lyophilized with 50 mM or 4% trehalose to prevent aggregation and stored at room temperature.
Collapse
Affiliation(s)
- William H. Fang
- Department of Orthopedic Surgery, Valley Health Systems, 620 Shadow Lane, Las Vegas, NV 89106, USA
| | - C. Thomas Vangsness
- Department of Orthopedic Surgery, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| |
Collapse
|
16
|
Daga KR, Larey AM, Morfin MG, Chen K, Bitarafan S, Carpenter JM, Hynds HM, Hines KM, Wood LB, Marklein RA. Microglia morphological response to mesenchymal stromal cell extracellular vesicles demonstrates EV therapeutic potential for modulating neuroinflammation. J Biol Eng 2024; 18:58. [PMID: 39420399 PMCID: PMC11488223 DOI: 10.1186/s13036-024-00449-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cell derived extracellular vesicles (MSC-EVs) are a promising therapeutic for neuroinflammation. MSC-EVs can interact with microglia, the resident immune cells of the brain, to exert their immunomodulatory effects. In response to inflammatory cues, such as cytokines, microglia undergo phenotypic changes indicative of their function e.g. morphology and secretion. However, these changes in response to MSC-EVs are not well understood. Additionally, no disease-relevant screening tools to assess MSC-EV bioactivity exist, which has further impeded clinical translation. Here, we developed a quantitative, high throughput morphological profiling approach to assess the response of microglia to neuroinflammation- relevant signals and whether this morphological response can be used to indicate the bioactivity of MSC-EVs. RESULTS Using an immortalized human microglia cell-line, we observed increased size (perimeter, major axis length) and complexity (form factor) upon stimulation with interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Upon treatment with MSC-EVs, the overall morphological score (determined using principal component analysis) shifted towards the unstimulated morphology, indicating that MSC-EVs are bioactive and modulate microglia. The morphological effects of MSC-EVs in TNF-α /IFN-γ stimulated cells were concomitant with reduced secretion of 14 chemokines/cytokines (e.g. CXCL6, CXCL9) and increased secretion of 12 chemokines/cytokines (e.g. CXCL8, CXCL10). Proteomic analysis of cell lysates revealed significant increases in 192 proteins (e.g. HIBADH, MEAK7, LAMC1) and decreases in 257 proteins (e.g. PTEN, TOM1, MFF) with MSC-EV treatment. Of note, many of these proteins are involved in regulation of cell morphology and migration. Gene Set Variation Analysis revealed upregulation of pathways associated with immune response, such as regulation of cytokine production, immune cell infiltration (e.g. T cells, NK cells) and morphological changes (e.g. Semaphorin, RHO/Rac signaling). Additionally, changes in microglia mitochondrial morphology were measured suggesting that MSC-EV modulate mitochondrial metabolism. CONCLUSION This study comprehensively demonstrates the effects of MSC-EVs on human microglial morphology, cytokine secretion, cellular proteome, and mitochondrial content. Our high-throughput, rapid, low-cost morphometric approach enables screening of MSC-EV batches and manufacturing conditions to enhance EV function and mitigate EV functional heterogeneity in a disease relevant manner. This approach is highly generalizable and can be further adapted and refined based on selection of the disease-relevant signal, target cell, and therapeutic product.
Collapse
Affiliation(s)
- Kanupriya R Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Andrew M Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G Morfin
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - Kailin Chen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Hannah M Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA.
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA.
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20903, USA.
| |
Collapse
|
17
|
Alsultan A, Farge D, Kili S, Forte M, Weiss DJ, Grignon F, Boelens JJ. International Society for Cell and Gene Therapy Clinical Translation Committee recommendations on mesenchymal stromal cells in graft-versus-host disease: easy manufacturing is faced with standardizing and commercialization challenges. Cytotherapy 2024; 26:1132-1140. [PMID: 38804990 PMCID: PMC12046531 DOI: 10.1016/j.jcyt.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Mesenchymal stromal cells (MSCs) have been used in multiple clinical trials for steroid-refractory moderate-severe (grade II-IV) acute graft-versus-host disease (aGVHD) across the world over the last two decades. Despite very promising results in a variety of trials, it failed to get widespread approval by regulatory agencies such as the U.S. Food and Drug Administration and the European Medicines Agency. What lessons can we learn from this for future studies on MSCs and other cell therapy products? Broad heterogeneity among published trials using MSCs in aGVHD was likely the core problem. We propose a standardized approach in regards to donor-related factors, MSCs-related characteristics, as well as clinical trial design, to limit heterogeneity in trials for aGVHD and to fulfill the requirements of regulatory agencies. This approach may be expanded beyond MSCs to other Cell and Gene therapy products and trials in other diseases.
Collapse
Affiliation(s)
- Abdulrahman Alsultan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Transplantation and Cellular Therapy, MSK Kids, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Dominique Farge
- Internal Medicine Unit (UF 04): CRMR MATHEC, Autoimmune diseases and Cellular Therapy, St-Louis Hospital, Center of reference for rare systemic autoimmune diseases of Ile-de-France (FAI2R), AP-HP, Hôpital St-Louis, Paris University, IRSL, Paris, France; Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sven Kili
- Sven Kili Consulting Ltd., Shrewsbury, UK; Saisei Ventures, Boston, Massachusetts, USA; CCRM, Toronto, Canada
| | | | - Daniel J Weiss
- University of Vermont College of Medicine, Burlington, Vermont, USA
| | - Felix Grignon
- International Society for Cell & Gene Therapy, Vancouver, Canada
| | - Jaap Jan Boelens
- Transplantation and Cellular Therapy, MSK Kids, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA.
| |
Collapse
|
18
|
Yin L, Ye M, Qiao Y, Huang W, Xu X, Xu S, Oh S. Unlocking the full potential of mesenchymal stromal cell therapy for osteoarthritis through machine learning-based in silico trials. Cytotherapy 2024; 26:1252-1263. [PMID: 38904585 DOI: 10.1016/j.jcyt.2024.05.016] [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: 01/29/2024] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024]
Abstract
Despite the potential of mesenchymal stromal cells (MSCs) in osteoarthritis (OA) treatment, the challenge lies in addressing their therapeutic inconsistency. Clinical trials revealed significantly varied therapeutic outcomes among patients receiving the same allogenic MSCs but different treatment regimens. Therefore, optimizing personalized treatment strategies is crucial to fully unlock MSCs' potential and enhance therapeutic consistency. We employed the XGBoost algorithm to train a self-collected database comprising 37 published clinical reports to create a model capable of predicting the probability of effective pain relief and Western Ontario and McMaster Universities (WOMAC) index improvement in OA patients undergoing MSC therapy. Leveraging this model, extensive in silico simulations were conducted to identify optimal personalized treatment strategies and ideal patient profiles. Our in silico trials predicted that the individually optimized MSC treatment strategies would substantially increase patients' chances of recovery compared to the strategies used in reported clinical trials, thereby potentially benefiting 78.1%, 47.8%, 94.4% and 36.4% of the patients with ineffective short-term pain relief, short-term WOMAC index improvement, long-term pain relief and long-term WOMAC index improvement, respectively. We further recommended guidelines on MSC number, concentration, and the patients' appropriate physical (body mass index, age, etc.) and disease states (Kellgren-Lawrence grade, etc.) for OA treatment. Additionally, we revealed the superior efficacy of MSC in providing short-term pain relief compared to platelet-rich plasma therapy for most OA patients. This study represents the pioneering effort to enhance the efficacy and consistency of MSC therapy through machine learning applied to clinical data. The in silico trial approach holds immense potential for diverse clinical applications.
Collapse
Affiliation(s)
- Lu Yin
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China; Agency for Science Technology and Research, Bioprocessing Technology Institute, Singapore, Singapore.
| | - Meiwu Ye
- Bio-totem Pte. Ltd., Guangzhou (Nanhai) Biomedical Industrial Park, Foshan, Guangdong, China
| | - Yang Qiao
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Weilu Huang
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Xinping Xu
- Jiangxi Provincial Key Laboratory of Respiratory Diseases, Jiangxi Institute of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Jiangxi Clinical Research Center for Respiratory Diseases, Nanchang, Jiangxi, China; Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, China
| | - Shuoyu Xu
- Bio-totem Pte. Ltd., Guangzhou (Nanhai) Biomedical Industrial Park, Foshan, Guangdong, China.
| | - Steve Oh
- Agency for Science Technology and Research, Bioprocessing Technology Institute, Singapore, Singapore; CellVec Pte. Ltd., Singapore, Singapore.
| |
Collapse
|
19
|
Wang J, Li R. Effects, methods and limits of the cryopreservation on mesenchymal stem cells. Stem Cell Res Ther 2024; 15:337. [PMID: 39343920 PMCID: PMC11441116 DOI: 10.1186/s13287-024-03954-3] [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] [Received: 06/19/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are a type of cell capable of regulating the immune system, as well as exhibiting self-renewal and multi-lineage differentiation potential. Mesenchymal stem cells have emerged as an essential source of seed cells for therapeutic cell therapy. It is crucial to cryopreserve MSCs in liquid nitrogen prior to clinical application while preserving their functionality. Furthermore, efficient cryopreservation greatly enhances MSCs' potential in a range of biological domains. Nevertheless, there are several limits on the MSC cryopreservation methods now in use, necessitating thorough biosafety assessments before utilizing cryopreserved MSCs. Therefore, in order to improve the effectiveness of cryopreserved MSCs in clinical stem cell treatment procedures, new technological techniques must be developed immediately. The study offers an exhaustive analysis of the state-of-the-art MSC cryopreservation techniques, their effects on MSCs, and the difficulties encountered when using cryopreserved MSCs in clinical applications.
Collapse
Affiliation(s)
- Jialing Wang
- Chengdu Senkicel Biotechnology Co. Ltd, Chengdu, China
| | - Rui Li
- Chengdu Senkicel Biotechnology Co. Ltd, Chengdu, China.
| |
Collapse
|
20
|
Chatzianagnosti S, Dermitzakis I, Theotokis P, Kousta E, Mastorakos G, Manthou ME. Application of Mesenchymal Stem Cells in Female Infertility Treatment: Protocols and Preliminary Results. Life (Basel) 2024; 14:1161. [PMID: 39337944 PMCID: PMC11433628 DOI: 10.3390/life14091161] [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: 07/22/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Infertility is a global phenomenon that impacts people of both the male and the female sex; it is related to multiple factors affecting an individual's overall systemic health. Recently, investigators have been using mesenchymal stem cell (MSC) therapy for female-fertility-related disorders such as polycystic ovarian syndrome (PCOS), premature ovarian failure (POF), endometriosis, preeclampsia, and Asherman syndrome (AS). Studies have shown promising results, indicating that MSCs can enhance ovarian function and restore fertility for affected individuals. Due to their regenerative effects and their participation in several paracrine pathways, MSCs can improve the fertility outcome. However, their beneficial effects are dependent on the methodologies and materials used from isolation to reimplantation. In this review, we provide an overview of the protocols and methods used in applications of MSCs. Moreover, we summarize the findings of published preclinical studies on infertility treatments and discuss the multiple properties of these studies, depending on the isolation source of the MSCs used.
Collapse
Affiliation(s)
- Sofia Chatzianagnosti
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Eleni Kousta
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George Mastorakos
- Department of Endocrinology, Diabetes Mellitus and Metabolism, Aretaieion Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
21
|
Faircloth TU, Temple S, Parr RN, Tucker AB, Rajan D, Hematti P, Kugathasan S, Chinnadurai R. Vascular endothelial growth factor secretion and immunosuppression are distinct potency mechanisms of human bone marrow mesenchymal stromal cells. Stem Cells 2024; 42:736-751. [PMID: 38826008 DOI: 10.1093/stmcls/sxae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
Mesenchymal stromal cells (MSCs) are investigated as cellular therapeutics for inflammatory bowel diseases and associated perianal fistula, although consistent efficacy remains a concern. Determining host factors that modulate MSCs' potency including their secretion of angiogenic and wound-healing factors, immunosuppression, and anti-inflammatory properties are important determinants of their functionality. We investigated the mechanisms that regulate the secretion of angiogenic and wound-healing factors and immune suppression of human bone marrow MSCs. Secretory analysis of MSCs focusing on 18 angiogenic and wound-healing secretory molecules identified the most abundancy of vascular endothelial growth factor A (VEGF-A). MSC viability and secretion of other angiogenic factors are not dependent on VEGF-A secretion which exclude the autocrine role of VEGF-A on MSC's fitness. However, the combination of inflammatory cytokines IFNγ and TNFα reduces MSC's VEGF-A secretion. To identify the effect of intestinal microvasculature on MSCs' potency, coculture analysis was performed between human large intestine microvascular endothelial cells (HLMVECs) and human bone marrow-derived MSCs. HLMVECs do not attenuate MSCs' viability despite blocking their VEGF-A secretion. In addition, HLMVECs neither attenuate MSC's IFNγ mediated upregulation of immunosuppressive enzyme indoleamine 2,3-dioxygenase nor abrogate suppression of T-cell proliferation despite the attenuation of VEGF-A secretion. We found that HLMVECs express copious amounts of endothelial nitric oxide synthase and mechanistic analysis showed that pharmacological blocking reverses HLMVEC-mediated attenuation of MSC's VEGF-A secretion. Together these results suggest that secretion of VEGF-A and immunosuppression are separable functions of MSCs which are regulated by distinct mechanisms in the host.
Collapse
Affiliation(s)
- Tyler U Faircloth
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| | - Sara Temple
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| | - Rhett N Parr
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| | - Anna B Tucker
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| | - Devi Rajan
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| | - Peiman Hematti
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, Atlanta, GA, United States
| | - Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31324, United States
| |
Collapse
|
22
|
Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products-a review. Front Cell Dev Biol 2024; 12:1400347. [PMID: 39129786 PMCID: PMC11310176 DOI: 10.3389/fcell.2024.1400347] [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: 03/13/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Mesenchymal stromal stem cells (MSCs) possess a remarkable potential for numerous clinical applications due to their unique properties including self-renewal, immunomodulation, paracrine actions and multilineage differentiation. However, the translation of MSC-based Advanced Therapy Medicinal Products (ATMPs) into the clinic has frequently met with inconsistent outcomes. One of the suspected reasons for this issue is the inherent and extensive variability that exists among such ATMPs, which makes the interpretation of their clinical efficacy difficult to assess, as well as to compare the results of various studies. This variability stems from numerous reasons including differences in tissue sources, donor attributes, variances in manufacturing protocols, as well as modes of administration. MSCs can be isolated from various tissues including bone marrow, umbilical cord, adipose tissue and others, each with its unique phenotypic and functional characteristics. While MSCs from different sources do share common features, they also exhibit distinct gene expression profiles and functional properites. Donor-specific factors such as age, sex, body mass index, and underlying health conditions can influence MSC phenotype, morphology, differentiation potential and function. Moreover, variations in preparation of MSC products introduces additional heterogeneity as a result of cell culture media composition, presence or absence of added growth factors, use of different serum supplements and culturing techniques. Once MSC products are formulated, storage protocols play a pivotal role in its efficacy. Factors that affect cell viability include cell concentration, delivery solution and importantly, post-thawing protocols where applicable. Ensuing, differences in administration protocols can critically affect the distribution and functionallity of administered cells. As MSC-based therapies continue to advance through numerous clinical trials, implication of strategies to reduce product heterogeneity is imperative. Central to addressing these challenges is the need for precise prediction of clinical responses, which require well-defined MSC populations and harmonized assessment of their specific functions. By addressing these issues by meaningful approaches, such as, e.g., MSC pooling, the field can overcome barriers to advance towards more consistent and effective MSC-based therapies.
Collapse
Affiliation(s)
- Ana Bajc Česnik
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
| | - Urban Švajger
- Slovenian Institute for Transfusion Medicine, Department for Therapeutic Services, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
23
|
Holland EN, Fernández-Yagüe MA, Zhou DW, O'Neill EB, Woodfolk AU, Mora-Boza A, Fu J, Schlaepfer DD, García AJ. FAK, vinculin, and talin control mechanosensitive YAP nuclear localization. Biomaterials 2024; 308:122542. [PMID: 38547833 PMCID: PMC11065566 DOI: 10.1016/j.biomaterials.2024.122542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/08/2024] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Abstract
Focal adhesions (FAs) are nanoscale complexes containing clustered integrin receptors and intracellular structural and signaling proteins that function as principal sites of mechanotransduction in part via promoting the nuclear translocation and activation of the transcriptional coactivator yes-associated protein (YAP). Knockdown of FA proteins such as focal adhesion kinase (FAK), talin, and vinculin can prevent YAP nuclear localization. However, the mechanism(s) of action remain poorly understood. Herein, we investigated the role of different functional domains in vinculin, talin, and FAK in regulating YAP nuclear localization. Using genetic or pharmacological inhibition of fibroblasts and human mesenchymal stem cells (hMSCs) adhering to deformable substrates, we find that disruption of vinculin-talin binding versus talin-FAK binding reduces YAP nuclear localization and transcriptional activity via different mechanisms. Disruption of vinculin-talin binding or knockdown of talin-1 reduces nuclear size, traction forces, and YAP nuclear localization. In contrast, disruption of the talin binding site on FAK or elimination of FAK catalytic activity did not alter nuclear size yet still prevented YAP nuclear localization and activity. These data support both nuclear tension-dependent and independent models for matrix stiffness-regulated YAP nuclear localization. Our results highlight the importance of vinculin-talin-FAK interactions at FAs of adherent cells, controlling YAP nuclear localization and activity.
Collapse
Affiliation(s)
- Elijah N Holland
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Marc A Fernández-Yagüe
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Department of Chemistry, Queen Mary University of London, London, UK
| | - Dennis W Zhou
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Eric B O'Neill
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ayanna U Woodfolk
- Mathematics Department, Spelman College, Atlanta, GA, USA; Bioengineering Department, North Carolina A&T State University, Greensboro, NC, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jianping Fu
- Department of Mechanical Engineering, Department of Biomedical Engineering, Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - David D Schlaepfer
- Moores Cancer Center, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
24
|
Chandanala S, Mohan G, Manukonda DL, Kumar A, Prasanna J. A novel role of CD73-IFNγ signalling axis in human mesenchymal stromal cell mediated inflammatory macrophage suppression. Regen Ther 2024; 26:89-101. [PMID: 38845846 PMCID: PMC11153905 DOI: 10.1016/j.reth.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 04/25/2024] [Accepted: 05/19/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction Immunomodulation is the predominant mechanism via which Mesenchymal stromal cells (MSCs) mediate their therapeutic benefits. However, inconsistent success in numerous clinical trials warrants a better understating of the molecular mechanisms regulating their immunomodulatory properties. CD73, an ecto-5'-nucleotidase is abundantly expressed by MSCs, however its precise role in regulating their immunomodulatory properties is still elusive. The present study explored the role of CD73 in Interferon-gamma (IFNγ) sensing and in turn their ability to suppress "inflammatory" M1 macrophages. Materials and methods CD73 knockdown MSCs (CD73-KDN) were initially assessed for expression of immunoregulatory molecules and IFNγ sensing ability by analysing expression of IFNγ signalling downstream targets such as pSTAT-1, Interferon-Stimulated Genes (ISG) and Indoleamine 2,3-dioxygnease (IDO), a prototypic IFNγ-induced immunomodulator. Next CD73-KDN MSCs were co-cultured with inflammatory M1 macrophages and evaluated for their ability to suppress them. To delineate the contributory role of CD73 and IFNγ signalling downstream target IDO, they were overexpressed independently in CD73-KDN MSCs and re-evaluated for their ability to suppress M1 macrophages. Results CD73-KDN MSCs exhibited reduced expression of immunoregulatory molecules and were refractory to IFNγ signalling as indicated by attenuated expression of pSTAT-1, Interferon-Stimulated Genes (ISG) and Indoleamine 2,3-dioxygnease (IDO) upon IFNγ exposure. Since sensing of inflammation is critical for MSC mediated immunomodulation, CD73-KDN MSCs were functionally evaluated for their ability to immune-modulate "inflammatory" M1 macrophages wherein they failed to suppress M1 macrophages. Interestingly, ectopic expression of either CD73 or IFNγ signalling target IDO1 in CD73-KDN MSCs restored their ability to suppress M1 macrophages, establishing the importance of CD73-IFNγ signalling axis in MSC-mediated inflammatory macrophage suppression. Conclusion The present study uncovers the unexplored role of CD73-IFNγ axis in MSC-mediated M1 macrophage suppression. MSC-educated macrophages are the actual immune-modulators at MSC transplant sites, thus CD73 can serve as a key immune-potency marker for benchmarking therapeutically relevant MSCs.
Collapse
Affiliation(s)
- Shashank Chandanala
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Govind Mohan
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - David-Luther Manukonda
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Anujith Kumar
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Jyothi Prasanna
- Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| |
Collapse
|
25
|
Chu W, Zhang F, Zeng X, He F, Shang G, Guo T, Wang Q, Wu J, Li T, Zhong ZZ, Liang X, Hu J, Liu M. A GMP-compliant manufacturing method for Wharton's jelly-derived mesenchymal stromal cells. Stem Cell Res Ther 2024; 15:131. [PMID: 38702793 PMCID: PMC11069138 DOI: 10.1186/s13287-024-03725-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) hold great therapeutic potential in regenerative medicine. Therefore, it is crucial to establish a Good Manufacturing Practice (GMP)-compliant methodology for the isolation and culture of WJ-MSCs. Through comprehensive research, encompassing laboratory-scale experiments to pilot-scale studies, we aimed to develop standardized protocols ensuring the high yield and quality of WJ-MSCs manufacturing. METHODS Firstly, optimization of parameters for the enzymatic digestion method used to isolate WJ-MSCs was conducted. These parameters included enzyme concentrations, digestion times, seeding densities, and culture media. Additionally, a comparative analysis between the explant method and the enzymatic digestion method was performed. Subsequently, the consecutive passaging of WJ-MSCs, specifically up to passage 9, was evaluated using the optimized method. Finally, manufacturing processes were developed and scaled up, starting from laboratory-scale flask-based production and progressing to pilot-scale cell factory-based production. Furthermore, a stability study was carried out to assess the storage and use of drug products (DPs). RESULTS The optimal parameters for the enzymatic digestion method were a concentration of 0.4 PZ U/mL Collagenase NB6 and a digestion time of 3 h, resulting in a higher yield of P0 WJ-MSCs. In addition, a positive correlation between the weight of umbilical cord tissue and the quantities of P0 WJ-MSCs has been observed. Evaluation of different concentrations of human platelet lysate revealed that 2% and 5% concentrations resulted in similar levels of cell expansion. Comparative analysis revealed that the enzymatic digestion method exhibited faster outgrowth of WJ-MSCs compared to the explant method during the initial passage. Passages 2 to 5 exhibited higher viability and proliferation ability throughout consecutive passaging. Moreover, scalable manufacturing processes from the laboratory scale to the pilot scale were successfully developed, ensuring the production of high-quality WJ-MSCs. Multiple freeze-thaw cycles of the DPs led to reduced cell viability and viable cell concentration. Subsequent thawing and dilution of the DPs resulted in a significant decrease in both metrics, especially when stored at 20-27 °C. CONCLUSION This study offers valuable insights into optimizing the isolation and culture of WJ-MSCs. Our scalable manufacturing processes facilitate the large-scale production of high-quality WJ-MSCs. These findings contribute to the advancement of WJ-MSCs-based therapies in regenerative medicine.
Collapse
Affiliation(s)
- Wanglong Chu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Fen Zhang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Xiuping Zeng
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Fangtao He
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Guanyan Shang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Tao Guo
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Qingfang Wang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Jianfu Wu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Tongjing Li
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Zhen Zhong Zhong
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Xiao Liang
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China
| | - Junyuan Hu
- Shenzhen Beike Biotechnology Co., Ltd, 518000, Shenzhen, Guangdong, People's Republic of China.
| | - Muyun Liu
- National Engineering Research Center of Foundational Technologies for CGT Industry, 518000, Shenzhen, Guangdong, People's Republic of China.
- Shenzhen Kenuo Medical Laboratory, 518000, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
26
|
Robb KP, Galipeau J, Shi Y, Schuster M, Martin I, Viswanathan S. Failure to launch commercially-approved mesenchymal stromal cell therapies: what's the path forward? Proceedings of the International Society for Cell & Gene Therapy (ISCT) Annual Meeting Roundtable held in May 2023, Palais des Congrès de Paris, Organized by the ISCT MSC Scientific Committee. Cytotherapy 2024; 26:413-417. [PMID: 37804284 DOI: 10.1016/j.jcyt.2023.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 09/02/2023] [Indexed: 10/09/2023]
Abstract
Mesenchymal stromal cells (MSCs) are promising cell therapy candidates, but their debated efficacy in clinical trials still limits successful adoption. Here, we discuss proceedings from a roundtable session titled "Failure to Launch Mesenchymal Stromal Cells 10 Years Later: What's on the Horizon?" held at the International Society for Cell & Gene Therapy 2023 Annual Meeting. Panelists discussed recent progress toward developing patient-stratification approaches for MSC treatments, highlighting the role of baseline levels of inflammation in mediating MSC treatment efficacy. In addition, MSC critical quality attributes (CQAs) are beginning to be elucidated and applied to investigational MSC products, including immunomodulatory functional assays and other potency markers that will help to ensure product consistency and quality. Lastly, next-generation MSC products, such as culture-priming strategies, were discussed as a promising strategy to augment MSC basal fitness and therapeutic potency. Key variables that will need to be considered alongside investigations of patient stratification approaches, CQAs and next-generation MSC products include the specific disease target being evaluated, route of administration of the cells and cell manufacturing parameters; these factors will have to be matched with postulated mechanisms of action towards treatment efficacy. Taken together, patient stratification metrics paired with the selection of therapeutically potent MSCs (using rigorous CQAs and/or engineered MSC products) represent a path forward to improve clinical successes and regulatory endorsements.
Collapse
Affiliation(s)
- Kevin P Robb
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jacques Galipeau
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin in Madison, Madison, Wisconsin, USA; University of Wisconsin Carbone Comprehensive Cancer, University of Wisconsin in Madison, Madison, Wisconsin, USA
| | - Yufang Shi
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; The Third Affiliated Hospital of Soochow University, The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University Medical College, Suzhou Jiangsu, China
| | | | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada; Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
27
|
Carlander ALF, Gundestrup AK, Jansson PM, Follin B, Hoeeg C, Kousholt BS, Larsen RT, Jakobsen KK, Rimborg S, Fischer-Nielsen A, Grønhøj C, Buchwald CV, Lynggaard CD. Mesenchymal Stromal/Stem Cell Therapy Improves Salivary Flow Rate in Radiation-Induced Salivary Gland Hypofunction in Preclinical in vivo Models: A Systematic Review and Meta-Analysis. Stem Cell Rev Rep 2024; 20:1078-1092. [PMID: 38430363 PMCID: PMC11087340 DOI: 10.1007/s12015-024-10700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Mesenchymal stromal/stem cells (MSCs) have been suggested for salivary gland (SG) restoration following radio-induced salivary gland damage. This study aimed to determine the safety and effectiveness of MSC therapy on radio-induced SG damage and hypofunction in preclinical in vivo studies. METHODS PubMed and EMBASE were systematically searched for preclinical in vivo interventional studies evaluating efficacy and safety of MSC treatment following radio-induced salivary gland damage published before 10th of January 2022. The primary endpoint was salivary flow rate (SFR) evaluated in a meta-analysis. The study protocol was published and registered on PROSPERO ( www.crd.ac.uk/prospero ), registration number CRD42021227336. RESULTS A total of 16 preclinical in vivo studies were included for qualitative analysis (858 experimental animals) and 13 in the meta-analysis (404 experimental animals). MSCs originated from bone marrow (four studies), adipose tissue (10 studies) and salivary gland tissue (two studies) and were administered intravenously (three studies), intra-glandularly (11 studies) or subcutaneously (one study). No serious adverse events were reported. The overall effect on SFR was significantly increased with a standardized mean difference (SMD) of 6.99 (95% CI: 2.55-11.42). Studies reported improvements in acinar tissue, vascular areas and paracrine factors. CONCLUSION In conclusion, this systematic review and meta-analysis showed a significant effect of MSC therapy for restoring SG functioning and regenerating SG tissue following radiotherapy in preclinical in vivo studies without serious adverse events. MSC therapy holds significant therapeutic potential in the treatment of radio-induced xerostomia, but comprehensive, randomized, clinical trials in humans are required to ascertain their efficacy in a clinical setting.
Collapse
Affiliation(s)
- Amanda-Louise Fenger Carlander
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
- Department of Otolaryngology, Head and Neck Surgery and Audiology, Rigshospitalet, Copenhagen University hospital, Copenhagen, Denmark.
| | - Anders Kierkegaard Gundestrup
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Per Marcus Jansson
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Cecilie Hoeeg
- Cardiology Stem Cell Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Birgitte Saima Kousholt
- Department of Clinical Medicine, Aarhus University Group for Understanding Systematic Reviews and Meta analyses in Translational Preclinical Science, Aarhus University, Copenhagen, Denmark
| | - Rasmus Tolstrup Larsen
- Department of Occupational Therapy and Physiotherapy, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Section of Social Medicine, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Kathrine Kronberg Jakobsen
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Susie Rimborg
- The Royal Danish Library, Copenhagen University Library, Copenhagen, Denmark
| | - Anne Fischer-Nielsen
- Department of Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Grønhøj
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian von Buchwald
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Duch Lynggaard
- Department of Otolaryngology and Audiology, Head and Neck Surgery, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
28
|
Sadeghi S, Nimtz L, Niebergall-Roth E, Norrick A, Hägele S, Vollmer L, Esterlechner J, Frank MH, Ganss C, Scharffetter-Kochanek K, Kluth MA. Potency assay to predict the anti-inflammatory capacity of a cell therapy product for macrophage-driven diseases: overcoming the challenges of assay development and validation. Cytotherapy 2024; 26:512-523. [PMID: 38441512 PMCID: PMC11065629 DOI: 10.1016/j.jcyt.2024.02.004] [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: 09/13/2023] [Revised: 01/22/2024] [Accepted: 02/12/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND Given the high level of product complexity and limited regulatory guidance, designing and implementing appropriate potency assays is often the most challenging part of establishing a quality control testing matrix for a cell-based medicinal product. Among the most elusive tasks are the selection of suitable read-out parameters, the development of assay designs that most closely model the pathophysiological conditions, and the validation of the methods. Here we describe these challenges and how they were addressed in developing an assay that measures the anti-inflammatory potency of mesenchymal stromal cells (MSCs) in an M1 macrophage-dominated inflammatory environment. METHODS An in vitro inflammation model was established by coculturing skin-derived ABCB5+ MSCs with THP-1 monocyte-derived M1-polarized macrophages. Readout was the amount of interleukin 1 receptor antagonist (IL-1RA) secreted by the MSCs in the coculture, measured by an enzyme-linked immunosorbent assay. RESULTS IL-1RA was quantified with guideline-concordant selectivity, accuracy and precision over a relevant concentration range. Consistent induction of the macrophage markers CD36 and CD80 indicated successful macrophage differentiation and M1 polarization of THP-1 cells, which was functionally confirmed by release of proinflammatory tumor necrosis factor α. Testing a wide range of MSC/macrophage ratios revealed the optimal ratio for near-maximal stimulation of MSCs to secrete IL-1RA, providing absolute maximum levels per individual MSC that can be used for future comparison with clinical efficacy. Batch release testing of 71 consecutively manufactured MSC batches showed a low overall failure rate and a high comparability between donors. CONCLUSIONS We describe the systematic development and validation of a therapeutically relevant, straightforward, robust and reproducible potency assay to measure the immunomodulatory capacity of MSCs in M1 macrophage-driven inflammation. The insights into the challenges and how they were addressed may also be helpful to developers of potency assays related to other cellular functions and clinical indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Markus H Frank
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA; Transplant Research Program, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA; School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | | | | | | |
Collapse
|
29
|
Klimczak A. Mesenchymal Stem/Progenitor Cells and Their Derivates in Tissue Regeneration-Part II. Int J Mol Sci 2024; 25:4937. [PMID: 38732156 PMCID: PMC11084558 DOI: 10.3390/ijms25094937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 05/13/2024] Open
Abstract
During the last three decades, mesenchymal stem/stromal cells (MSCs) were extensively studied, and are mainly considered within the setting of their regenerative and immunomodulatory properties in tissue regeneration [...].
Collapse
Affiliation(s)
- Aleksandra Klimczak
- Laboratory of Biology of Stem and Neoplastic Cells, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, R. Weigla 12, 53-114 Wroclaw, Poland
| |
Collapse
|
30
|
Blitzer GC, Paz C, Glassey A, Ganz OR, Giri J, Pennati A, Meyers RO, Bates AM, Nickel KP, Weiss M, Morris ZS, Mattison RJ, McDowell KA, Croxford E, Chappell RJ, Glazer TA, Rogus-Pulia NM, Galipeau J, Kimple RJ. Functionality of bone marrow mesenchymal stromal cells derived from head and neck cancer patients - A FDA-IND enabling study regarding MSC-based treatments for radiation-induced xerostomia. Radiother Oncol 2024; 192:110093. [PMID: 38224919 PMCID: PMC10922976 DOI: 10.1016/j.radonc.2024.110093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/17/2024]
Abstract
PURPOSE Salivary dysfunction is a significant side effect of radiation therapy for head and neck cancer (HNC). Preliminary data suggests that mesenchymal stromal cells (MSCs) can improve salivary function. Whether MSCs from HNC patients who have completed chemoradiation are functionally similar to those from healthy patients is unknown. We performed a pilot clinical study to determine whether bone marrow-derived MSCs [MSC(M)] from HNC patients could be used for the treatment of RT-induced salivary dysfunction. METHODS An IRB-approved pilot clinical study was undertaken on HNC patients with xerostomia who had completed treatment two or more years prior. Patients underwent iliac crest bone marrow aspirate and MSC(M) were isolated and cultured. Culture-expanded MSC(M) were stimulated with IFNγ and cryopreserved prior to reanimation and profiling for functional markers by flow cytometry and ELISA. MSC(M) were additionally injected into mice with radiation-induced xerostomia and the changes in salivary gland histology and salivary production were examined. RESULTS A total of six subjects were enrolled. MSC(M) from all subjects were culture expanded to > 20 million cells in a median of 15.5 days (range 8-20 days). Flow cytometry confirmed that cultured cells from HNC patients were MSC(M). Functional flow cytometry demonstrated that these IFNγ-stimulated MSC(M) acquired an immunosuppressive phenotype. IFNγ-stimulated MSC(M) from HNC patients were found to express GDNF, WNT1, and R-spondin 1 as well as pro-angiogenesis and immunomodulatory cytokines. In mice, IFNγ-stimulated MSC(M) injection after radiation decreased the loss of acinar cells, decreased the formation of fibrosis, and increased salivary production. CONCLUSIONS MSC (M) from previously treated HNC patients can be expanded for auto-transplantation and are functionally active. Furthermore IFNγ-stimulated MSC(M) express proteins implicated in salivary gland regeneration. This study provides preliminary data supporting the feasibility of using autologous MSC(M) from HNC patients to treat RT-induced salivary dysfunction.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Cristina Paz
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Annemarie Glassey
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Olga R Ganz
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Jayeeta Giri
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Andrea Pennati
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ross O Meyers
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Amber M Bates
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kwangok P Nickel
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Marissa Weiss
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Zachary S Morris
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Ryan J Mattison
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Kimberly A McDowell
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Emma Croxford
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA
| | - Richard J Chappell
- Department of Biostatistics and Medical Informatics, 610 Walnut Street, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53726 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Tiffany A Glazer
- Department of Surgery, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Nicole M Rogus-Pulia
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; Geriatric Research Education and Clinical Center, 2500 Overlook Terrace, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705 USA
| | - Jacques Galipeau
- Department of Medicine, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA
| | - Randall J Kimple
- Department of Human Oncology, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA; UW Carbone Cancer Center, 600 Highland Ave, University of Wisconsin, School of Medicine and Public Health, Madison, WI 53705 USA.
| |
Collapse
|
31
|
Pereira L, Echarte L, Romero M, Grazioli G, Pérez-Campos H, Francia A, Vicentino W, Mombrú AW, Faccio R, Álvarez I, Touriño C, Pardo H. Synthesis and characterization of a bovine collagen: GAG scaffold with Uruguayan raw material for tissue engineering. Cell Tissue Bank 2024; 25:123-142. [PMID: 34536180 DOI: 10.1007/s10561-021-09960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
Tissue engineering (TE) and regenerative medicine offer strategies to improve damaged tissues by using scaffolds and cells. The use of collagen-based biomaterials in the field of TE has been intensively growing over the past decades. Mesenchymal stromal cells (MSCs) and dental pulp stem cells (DPSCs) are promising cell candidates for development of clinical composites. In this study, we proposed the development of a bovine collagen type I: chondroitin-6-sulphate (CG) scaffold, obtained from Uruguayan raw material (certified as free bovine spongiform encephalopathy), with CG crosslinking enhancement using different gamma radiation doses. Structural, biomechanical and chemical characteristics of the scaffolds were assessed by Scanning Electron Microscopy, axial tensile tests, FT-IR and Raman Spectroscopy, respectively. Once we selected the most appropriate scaffold for future use as a TE product, we studied the behavior of MSCs and DPSCs cultured on the scaffold by cytotoxicity, proliferation and differentiation assays. Among the diverse porous scaffolds obtained, the one with the most adequate properties was the one exposed to 15 kGy of gamma radiation. This radiation dose contributed to the crosslinking of molecules, to the formation of new bonds and/or to the reorganization of the collagen fibers. The selected scaffold was non-cytotoxic for the tested cells and a suitable substrate for cell proliferation. Furthermore, the scaffold allowed MSCs differentiation to osteogenic, chondrogenic, and adipogenic lineages. Thus, this work shows a promising approach to the synthesis of a collagen-scaffold suitable for TE.
Collapse
Affiliation(s)
- L Pereira
- Centro NanoMat, Facultad de Química, Instituto Polo Tecnológico de Pando, UdelaR, Camino Aparicio Saravia s/n, 9100, Pando, Canelones, Uruguay
| | - L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - M Romero
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - H Pérez-Campos
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A Francia
- Fisiología general y bucodental, Facultad de Odontología, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - A W Mombrú
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - R Faccio
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay
| | - I Álvarez
- Instituto Nacional de Donación y Trasplante (INDT), Ministerio de salud Pública-Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Ministerio, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, Universidad de la República (UdelaR), Montevideo, Uruguay.
| | - H Pardo
- Cátedra de Física, Facultad de Química, DETEMA, Universidad de la República (UdelaR), General Flores, 2124, 11800, Montevideo, Uruguay.
| |
Collapse
|
32
|
Greco R, Alexander T, Del Papa N, Müller F, Saccardi R, Sanchez-Guijo F, Schett G, Sharrack B, Snowden JA, Tarte K, Onida F, Sánchez-Ortega I, Burman J, Castilla Llorente C, Cervera R, Ciceri F, Doria A, Henes J, Lindsay J, Mackensen A, Muraro PA, Ricart E, Rovira M, Zuckerman T, Yakoub-Agha I, Farge D. Innovative cellular therapies for autoimmune diseases: expert-based position statement and clinical practice recommendations from the EBMT practice harmonization and guidelines committee. EClinicalMedicine 2024; 69:102476. [PMID: 38361991 PMCID: PMC10867419 DOI: 10.1016/j.eclinm.2024.102476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/05/2024] [Accepted: 01/24/2024] [Indexed: 02/17/2024] Open
Abstract
Autoimmune diseases (ADs) are characterized by loss of immune tolerance, high chronicity, with substantial morbidity and mortality, despite conventional immunosuppression (IS) or targeted disease modifying therapies (DMTs), which usually require repeated administration. Recently, novel cellular therapies (CT), including mesenchymal stromal cells (MSC), Chimeric Antigen Receptors T cells (CART) and regulatory T cells (Tregs), have been successfully adopted in ADs. An international expert panel of the European Society for Blood and Marrow Transplantation and the International Society for the Cell and Gene Therapy, reviewed all available evidence, based on the current literature and expert practices, on use of MSC, CART and Tregs, in AD patients with rheumatological, neurological, and gastroenterological indications. Expert-based consensus and recommendations for best practice and quality of patient care were developed to support clinicians, scientists, and their multidisciplinary teams, as well as patients and care providers and will be regularly updated.
Collapse
Affiliation(s)
- Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT and Chair of the ADWP of the EBMT, Barcelona, Spain
| | - Tobias Alexander
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Nicoletta Del Papa
- Scleroderma Clinic, Rheumatology Department, ASST G. Pini-CTO, Università degli Studi di Milano, Milano, Italy
| | - Fabian Müller
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Riccardo Saccardi
- Cellular Therapies and Transfusion Medicine Unit, Careggi University Hospital, Florence, Italy
| | - Fermin Sanchez-Guijo
- Department of Hematology, IBSAL-University Hospital of Salamanca and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, FAU Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Universitätsklinikum Erlangen, Friedrich-Alexander University (FAU) Erlangen- Nürnberg, Erlangen, Germany
| | - Basil Sharrack
- Department of Neuroscience and Sheffield NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals NHS Foundation Trust & University of Sheffield, Sheffield, England, United Kingdom
| | - John A. Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, UK
| | - Karin Tarte
- SITI Lab, CHU Rennes, EFS Bretagne, University Rennes, Rennes, France
| | - Francesco Onida
- Hematology & ASCT Unit, ASST Fatebenefratelli-Sacco, University of Milan, Italy
- Co-Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Isabel Sánchez-Ortega
- Secretary of the Practice Harmonization and Guidelines Committee of EBMT, Barcelona, Spain
- EBMT Medical Officer, Executive Office, Barcelona, Spain
| | - Joachim Burman
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Ricard Cervera
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC, CSUR) of the Catalan and Spanish Health Systems/Member of ERN-ReCONNET, Hospital Clínic, Barcelona, Catalonia, Spain
| | - Fabio Ciceri
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea Doria
- Rheumatology Unit, Department of Medicine (DiMED), University of Padua, Padua, Italy
| | - Jörg Henes
- Center for Interdisciplinary Rheumatology, Immunology and Autoimmune diseases and Department of Internal Medicine II (Haematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Germany
| | - James Lindsay
- Department of Gastroenterology, The Royal London Hospital, Barts Health NHS Trust, London, UK
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology and Oncology, University Hospital of Erlangen, Erlangen, Germany
- Bayrisches Zentrum für Krebsforschung (BZKF) Erlangen, Germany
| | - Paolo A. Muraro
- Department of Brain Sciences, Imperial College London, London, UK
| | - Elena Ricart
- Gastroenterology Department. Hospital Clínic Barcelona. Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS). Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Montserrat Rovira
- BMT Unit, Haematology Department, Institute of Haematology and Oncology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Foundation, Spain
| | - Tsila Zuckerman
- Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ibrahim Yakoub-Agha
- CHU de Lille, University Lille, INSERM U1286, Infinite, 59000, Lille, France
- Chair of the Practice Harmonization and Guidelines Committee of EBMT, Spain
| | - Dominique Farge
- Internal Medicine Unit (04): CRMR MATHEC, Maladies Auto-immunes et Thérapie Cellulaire, Centre de Référence des Maladies auto-immunes systémiques Rares d’Ile-de-France, AP-HP, St-Louis Hospital Paris-Cite University, France
- Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
33
|
Karami Fath M, Bagherzadeh Torbati SM, Saqagandomabadi V, Yousefi Afshar O, Khalilzad M, Abedi S, Moliani A, Daneshdoust D, Barati G. The therapeutic effect of MSCs and their extracellular vesicles on neuroblastoma. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 187:51-60. [PMID: 38373516 DOI: 10.1016/j.pbiomolbio.2024.02.004] [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: 05/29/2023] [Revised: 10/04/2023] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Neuroblastoma is a common inflammatory-related cancer during infancy. Standard treatment modalities including surgical interventions, high-dose chemotherapy, radiotherapy, and immunotherapy are not able to increase survival rate and reduce tumor relapse in high-risk patients. Mesenchymal stem cells (MSCs) are known for their tumor-targeting and immunomodulating properties. MSCs could be engineered to express anticancer agents (i.e., growth factors, cytokines, pro-apoptotic agents) or deliver oncolytic viruses in the tumor microenvironment. As many functions of MSCs are mediated through their secretome, researchers have tried to use extracellular vesicles (EVs) from MSCs for targeted therapy of neuroblastoma. Here, we reviewed the studies to figure out whether the use of MSCs could be worthwhile in neuroblastoma therapy or not. Native MSCs have shown a promoting or inhibiting role in cancers including neuroblastoma. Therefore, MSCs are proposed as a vehicle to deliver anticancer agents such as oncolytic viruses to the neuroblastoma tumor microenvironment. Although modified MSCs or their EVs have been shown to suppress the tumorigenesis of neuroblastoma, further pre-clinical and clinical studies are required to come to a conclusion.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Vahid Saqagandomabadi
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Mohammad Khalilzad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sara Abedi
- Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Afshin Moliani
- Isfahan Medical Students Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Danyal Daneshdoust
- Faculty of Medicine, Babol University of Medical Sciences, Mazandaran, Iran
| | | |
Collapse
|
34
|
Echarte L, Grazioli G, Pereira L, Francia A, Pérez H, Kuzuian W, Vicentino W, Pardo H, Mombrú A, Maglia Á, Touriño C, Álvarez I. Processing methods for human amniotic membrane as scaffold for tissue engineering with mesenchymal stromal human cells. Cell Tissue Bank 2024; 25:269-283. [PMID: 35906514 DOI: 10.1007/s10561-022-10014-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 04/27/2022] [Indexed: 11/30/2022]
Abstract
Tissue engineering is an interdisciplinary field that applies the principles of engineering and life sciences toward the development of biological substitutes that restore, maintain, or improve tissue function. The aims of this work were to compare chemically and physically processed human Amniotic Membranes (hAM) and analyze the cytocompatibility and proliferation rate (PR) of two primary human mesenchymal stromal cell lines, from different sources and donor conditions seeded over these scaffolds. The evaluated hAM processes were: cold shock to obtain a frozen amniotic membrane (FEAM) with remaining dead epithelial cells, denudation of hAM with trypsin for 20/10 min (DEAM20/10) or treatment with sodium dodecyl sulfate to decellularized hAM (DAM). All samples were sterilized with gamma radiation. The selection of the treated hAM to then generate composites was performed by scanning and transmission electron microscopy and characterization by X-ray diffraction, selecting DEAM10 and FEAM as scaffolds for cell seeding. Two sources of primary human stromal cells were used, both developed by our researchers, human Dental Pulp Stem Cells (hDPSC) from living donors and human Mesenchymal Stromal Cells (hMSC) from bone marrow isolated from brain dead donors. This last line of cells conveys a novel source of human cells that, to our knowledge, have not been tested as part of this type of construct. We developed four in vitro constructs without cytotoxicity signs and with different PR depending on the scaffolds and cells. hDPSC and hMSC grew over both FEAM and DEAM10, but DEAM10 allowed higher PR.
Collapse
Affiliation(s)
- L Echarte
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - G Grazioli
- Cátedra de Materiales Dentales, Facultad de Odontología UdelaR, Montevideo, Uruguay
| | - L Pereira
- Departamento de Biomateriales, Facultad de Química, Parque Científico Tecnológico de Pando, UdelaR, Canelones, Uruguay
| | - A Francia
- Facultad de Odontología UdelaR, Fisiología General y Bucodental, Montevideo, Uruguay
| | - H Pérez
- Facultad de Medicina, Instituto Nacional de Donación y Trasplante (INDT), Ministerio de Salud Pública- Hospital de Clínicas, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - W Kuzuian
- Facultad de Medicina, Instituto Nacional de Donación y Trasplante (INDT), Ministerio de Salud Pública- Hospital de Clínicas, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - W Vicentino
- Facultad de Medicina, Instituto Nacional de Donación y Trasplante (INDT), Ministerio de Salud Pública- Hospital de Clínicas, Universidad de La República (UdelaR), Montevideo, Uruguay
| | - H Pardo
- Departamento de Biomateriales, Facultad de Química, Parque Científico Tecnológico de Pando, UdelaR, Canelones, Uruguay
| | - A Mombrú
- Departamento de Biomateriales, Facultad de Química, Parque Científico Tecnológico de Pando, UdelaR, Canelones, Uruguay
| | - Á Maglia
- Facultad de Odontología UdelaR, Cátedra de Histología y Embriología Bucodental, Montevideo, Uruguay
| | - C Touriño
- Área Terapia Celular y Medicina Regenerativa (ATCMR), Departamento Básico de Medicina, Hospital de Clínicas, Facultad de Medicina, UdelaR, Montevideo, Uruguay
| | - I Álvarez
- Facultad de Medicina, Instituto Nacional de Donación y Trasplante (INDT), Ministerio de Salud Pública- Hospital de Clínicas, Universidad de La República (UdelaR), Montevideo, Uruguay.
| |
Collapse
|
35
|
Galera MR, Svalgaard J, Woetmann A. Therapeutic potential of adipose derived stromal cells for major skin inflammatory diseases. Front Med (Lausanne) 2024; 11:1298229. [PMID: 38463491 PMCID: PMC10921940 DOI: 10.3389/fmed.2024.1298229] [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: 09/21/2023] [Accepted: 01/31/2024] [Indexed: 03/12/2024] Open
Abstract
Inflammatory skin diseases like psoriasis and atopic dermatitis are chronic inflammatory skin conditions continuously under investigation due to increased prevalence and lack of cure. Moreover, long-term treatments available are often associated with adverse effects and drug resistance. Consequently, there is a clear unmet need for new therapeutic approaches. One promising and cutting-edge treatment option is the use of adipose-derived mesenchymal stromal cells (AD-MSCs) due to its immunomodulatory and anti-inflammatory properties. Therefore, this mini review aims to highlight why adipose-derived mesenchymal stromal cells are a potential new treatment for these diseases by summarizing the pre-clinical and clinical studies investigated up to date and addressing current limitations and unresolved clinical questions from a dermatological and immunomodulatory point of view.
Collapse
Affiliation(s)
- Marina Ramírez Galera
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Anders Woetmann
- The LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
36
|
Nguyen Thi YV, Ngo AD, Chu DT, Lin SC, Wu CC. RNA therapeutics for regenerative medicine. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 204:163-176. [PMID: 38458737 DOI: 10.1016/bs.pmbts.2023.12.002] [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: 03/10/2024]
Abstract
It is estimated that millions of people around the world experience various types of tissue injuries every year. Regenerative medicine was born and developed for understanding and application with the aim of replacing affected organs or some cells. The research, manufacture, production, and distribution of RNA in cells have acted as a basic foundation for the development and testing of therapies and treatments that are widely applied in different fields of medicine. Vaccines against COVID-19 are considered one of the brilliant and outstanding successes of RNA therapeutics research. With the characteristics of bio-derived RNA therapeutics, the mechanism of rapid implementation, safe production, and flexibility to create proteins depending on actual requirements. Based on the advantages above in this review, we discuss RNA therapeutics for regenerative medicine, and the types of RNA therapies currently being used for regenerative medicine. The relationship between disease and regenerative medicine is currently being studied or tested in RNA therapeutics. We have also covered the mechanisms of action of RNA therapy for regenerative medicine and some of the limitations in our current understanding of the effects of RNA therapy in this area. Additionally, we have also covered developing RNA therapeutics for regenerative medicine, focusing on RNA therapeutics for regenerative medicine. As a final point, we discuss potential applications for therapeutics for regenerative medicine in the future, as well as their mechanisms.
Collapse
Affiliation(s)
- Yen Vy Nguyen Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Anh Dao Ngo
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Sheng-Che Lin
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Plastic and Reconstructive Surgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan.
| | - Chia-Ching Wu
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan; Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
37
|
Ataei A, Azizi M, Hajisadeghi S, Madani M, Khorami M, Hassantash S, Saeidpour Masouleh S, Barati G. The Therapeutic Effects of Mesenchymal Stem Cells and their Secretome on Oral Squamous Cell Carcinoma. Curr Mol Med 2024; 24:1195-1207. [PMID: 37366360 DOI: 10.2174/1566524023666230627151809] [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: 10/27/2022] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023]
Abstract
Oral cancers are prevalent in the human population, particularly in unindustrialized countries. In 90 % of oral cancers, the tumors arise from squamous cells, which is called oral squamous cell carcinoma (OSCC). Despite new treatment strategies, the morbidity and mortality rates are still high. Current treatment options including surgery, chemotherapy, and radiotherapy are not effective in the treatment of the tumor. Cell therapy with mesenchymal stem cells (MSCs) is considered one of the leading strategies in cancer treatment. However, the field of MSC therapy in OSCC is immature and ongoing studies are being conducted in experimental and pre-clinical studies. Here, we reviewed these studies to figure out whether the use of MSCs could be worthwhile in OSCC therapy or not. Both native and engineered MSCs as well as their secretome have been used in the treatment of OSCC. It seems that genetically modified MSCs or their secretome could inhibit the tumorigenesis of OSCC. However, further pre-clinical studies are required to come to a conclusion.
Collapse
Affiliation(s)
- Atefe Ataei
- Department of Periodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Azizi
- Department of Periodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | - Samira Hajisadeghi
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Qom University of Medical Sciences, Qom, Iran
| | - Mojan Madani
- Orthodontics Department, Dental Faculty, Arak UNDUniversity of Medical Sciences, Arak, Iran
| | - Mozhgan Khorami
- Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Hassantash
- Faculty of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ghasem Barati
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Stem Cell Technology Research Center, Tehran, Iran
| |
Collapse
|
38
|
Wang X, Wang Q, Meng L, Tian R, Guo H, Tan Z, Tan Y. Biodistribution-based Administration of cGMP-compliant Human Umbilical Cord Mesenchymal Stem Cells Affects the Therapeutic Effect of Wound Healing. Stem Cell Rev Rep 2024; 20:329-346. [PMID: 37889447 DOI: 10.1007/s12015-023-10644-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Although mesenchymal stem cells (MSCs) are used as therapeutic agents for skin injury therapy, few studies have reported the effects of dosing duration and delivery frequency on wound healing. In addition, before the clinical application of MSCs, it is important to assess whether their usage might influence tumor occurrence. METHODS We described the metabolic patterns of subcutaneous injection of hUC-MSCs using fluorescence tracing and qPCR methods and applied them to the development of drug delivery strategies for promoting wound healing. RESULTS (i) We developed cGMP-compliant hUC-MSC products with critical quality control points for wound healing; (ii) The products did not possess any tumorigenic or tumor-promoting/inhibiting ability in vivo; (iii) Fluorescence tracing and qPCR analyses showed that the subcutaneous application of hUC-MSCs did not result in safety-relevant biodistribution or ectopic migration; (iv) Reinjecting hUC-MSCs after significant consumption significantly improved reepithelialization and dermal regeneration. CONCLUSIONS Our findings provided a reference for controlling the quality of MSC products used for wound healing and highlighted the importance of delivery time and frequency for designing in vivo therapeutic studies.
Collapse
Affiliation(s)
- Xin Wang
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Qiuhong Wang
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Lingjiao Meng
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Ruifeng Tian
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Huizhen Guo
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China
| | - Zengqi Tan
- School of Medicine, Northwest University, Xi'an, China
| | - Yi Tan
- Qilu Cell Therapy Technology Co., Ltd, Gangyuan 6th Road, Licheng District, Ji'nan, Shandong, 250000, People's Republic of China.
- Shandong Yinfeng Life Science Research Institute, Ji'nan, People's Republic of China.
| |
Collapse
|
39
|
Nejabati HR, Nikzad S, Roshangar L. Therapeutic Potential of Mesenchymal Stem Cells in PCOS. Curr Stem Cell Res Ther 2024; 19:134-144. [PMID: 37198984 DOI: 10.2174/1574888x18666230517123256] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a major reproductive endocrine disorder affecting different facets of a woman's life, comprising reproduction, metabolism, and mental health. Recently, several research groups have brought attention to the therapeutic capacity of mesenchymal stem cells (MSCs) for the treatment of female reproductive disorders. It is highlighted that the treatment with bone marrow mesenchymal stem cells (BMMSCs) considerably diminishes the levels of some inflammatory markers as well as essential genes for ovarian production of androgens, which are considerably higher in theca cells of PCOS women than in those of healthy cases. In addition, studies show that BMMSCs improve in vitro maturation (IVM) of germinal vesicles (GVs) and the number of antral follicles while lessening the number of primary and preantral follicles in mice with PCOS compared to healthy controls. Regarding adipose- derived mesenchymal stem cells (AdMSCs), these cells restore the ovarian structure, enhance the number of oocytes and corpora luteum, and diminish the number of aberrant cystic follicles in PCOS rats. Some research also indicates that umbilical cord mesenchymal stem cells (UC-MSCs) alleviate the inflammation of granulosa cells in women with PCOS. Therefore, due to the limited research on MSC therapy in PCOS, in this review, we summarize the current knowledge on the therapeutic potential of three types of MSCs: BMMSCs, AdMSCs, UC-MSCs and their secretome in the treatment of PCOS.
Collapse
Affiliation(s)
- Hamid Reza Nejabati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadeneh Nikzad
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
40
|
Harland N, Knoll J, Amend B, Abruzzese T, Abele H, Jakubowski P, Stenzl A, Aicher WK. Xenogenic Application of Human Placenta-Derived Mesenchymal Stromal Cells in a Porcine Large Animal Model. Cell Transplant 2024; 33:9636897241226737. [PMID: 38323325 PMCID: PMC10851762 DOI: 10.1177/09636897241226737] [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] [Received: 07/31/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
In animal models, cell therapies for different diseases or injuries have been very successful. Preclinical studies with cells aiming at a stroke, heart attack, and other emergency situations were promising but sometimes failed translation in clinical situations. We, therefore, investigated if human placenta-derived mesenchymal stromal cells can be injected in pigs without provoking rejection to serve as a xenogenic transplantation model to bridge preclinical animal studies to more promising future preclinical studies. Male human placenta-derived mesenchymal stromal cells were isolated, expanded, and characterized by flow cytometry, in vitro differentiation, and quantitative reverse-transcription polymerase chain reaction to prove their nature. Such cells were injected into the sphincter muscle of the urethrae of female pigs under visual control by cystoscopy employing a Williams needle. The animals were observed over 7 days of follow-up. Reactions of the host to the xenogeneic cells were explored by monitoring body temperature, and inflammatory markers including IL-1ß, CRP, and haptoglobin in blood. After sacrifice on day 7, infiltration of inflammatory cells in the tissue targeted was investigated by histology and immunofluorescence. DNA of injected human cells was detected by PCR. Upon injection in vascularized porcine tissue, human placenta-derived mesenchymal stromal cells were tolerated, and systemic inflammatory parameters were not elevated. DNA of injected cells was detected in situ 7 days after injection, and moderate local infiltration of inflammatory cells was observed. The therapeutic potential of human placenta-derived mesenchymal stromal cells can be explored in porcine large animal models of injury or disease. This seems a promising strategy to explore technologies for cell injections in infarcted hearts or small organs and tissues in therapeutically relevant amounts requiring large animal models to yield meaningful outcomes.
Collapse
Affiliation(s)
- Niklas Harland
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Jasmin Knoll
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Bastian Amend
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Tanja Abruzzese
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Harald Abele
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Peter Jakubowski
- Department of Gynecology and Obstetrics, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Arnulf Stenzl
- Center for Medical Research, University Hospital, Eberhard Karls University, Tuebingen, Germany
| | - Wilhelm K. Aicher
- Department of Urology, University Hospital, Eberhard Karls University, Tuebingen, Germany
| |
Collapse
|
41
|
Yang H, Cheong S, He Y, Lu F. Mesenchymal stem cell-based therapy for autoimmune-related fibrotic skin diseases-systemic sclerosis and sclerodermatous graft-versus-host disease. Stem Cell Res Ther 2023; 14:372. [PMID: 38111001 PMCID: PMC10729330 DOI: 10.1186/s13287-023-03543-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/23/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) and sclerodermatous graft-versus-host disease (Scl-GVHD)-characterized by similar developmental fibrosis, vascular abnormalities, and innate and adaptive immune response, resulting in severe skin fibrosis at the late stage-are chronic autoimmune diseases of connective tissue. The significant immune system dysfunction, distinguishing autoimmune-related fibrosis from mere skin fibrosis, should be a particular focus of treating autoimmune-related fibrosis. Recent research shows that innovative mesenchymal stem cell (MSC)-based therapy, with the capacities of immune regulation, inflammation suppression, oxidation inhibition, and fibrosis restraint, shows great promise in overcoming the disease. MAIN BODY This review of recent studies aims to summarize the therapeutic effect and theoretical mechanisms of MSC-based therapy in treating autoimmune-related fibrotic skin diseases, SSc and Scl-GVHD, providing novel insights and references for further clinical applications. It is noteworthy that the efficacy of MSCs is not reliant on their migration into the skin. Working on the immune system, MSCs can inhibit the chemotaxis and infiltration of immune cells to the skin by down-regulating the expression of skin chemokines and chemokine receptors and reducing the inflammatory and pro-fibrotic mediators. Furthermore, to reduce levels of oxidative stress, MSCs may improve vascular abnormalities, and enhance the antioxidant defenses through inducible nitric oxide synthase, thioredoxin 1, as well as other mediators. The oxidative stress environment does not weaken MSCs and may even strengthen certain functions. Regarding fibrosis, MSCs primarily target the transforming growth factor-β signaling pathway to inhibit fibroblast activation. Here, miRNAs may play a critical role in ECM remodeling. Clinical studies have demonstrated the safety of these approaches, though outcomes have varied, possibly owing to the heterogeneity of MSCs, the disorders themselves, and other factors. Nevertheless, the research clearly reveals the immense potential of MSCs in treating autoimmune-related fibrotic skin diseases. CONCLUSION The application of MSCs presents a promising approach for treating autoimmune-related fibrotic skin diseases: SSc and Scl-GVHD. Therapies involving MSCs and MSC extracellular vesicles have been found to operate through three primary mechanisms: rebalancing the immune and inflammatory disorders, resisting oxidant stress, and inhibiting overactivated fibrosis (including fibroblast activation and ECM remodeling). However, the effectiveness of these interventions requires further validation through extensive clinical investigations, particularly randomized control trials and phase III/IV clinical trials. Additionally, the hypothetical mechanism underlying these therapies could be elucidated through further research.
Collapse
Affiliation(s)
- Han Yang
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Sousan Cheong
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yunfan He
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| | - Feng Lu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
42
|
Delgadillo J, Kerkelä E, Waters A, Akker EVD, Lechanteur C, Baudoux E, Gardiner N, De Vos J, Vives J. A management model in blood, tissue and cell establishments to ensure rapid and sustainable patient access to advanced therapy medicinal products in Europe. Cytotherapy 2023; 25:1259-1264. [PMID: 37737767 DOI: 10.1016/j.jcyt.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/23/2023]
Abstract
Blood, tissue and cell establishments (BTCs) stand out in the management of donor selection, procurement and processing of all types of substances of human origin (SoHO). In the last decades, the framework created around BTCs, including hospitals and national health system networks, and their links to research, development and innovation organizations and agencies have spurred their involvement in the study of groundbreaking advanced therapy medicinal products (ATMP). To further improve strategic synergies in the development of ATMPs, it will be required to promote intra- and inter-European collaborations by creating an international network involving BTCs and major stakeholders (i.e., research organizations, hospitals, universities, patient associations, public agencies). This vision is already shared with the European Blood Alliance, the association of non-profit blood establishments, with 26 member states throughout the European Union and European Free Trade Association states. Herein we present and analyze the "BTC for ATMP Development And Manufacture" (BADAM) model, an ethically responsible business model based on the values and missions of BTCs and their commitment to health equity, patient access and education (based on voluntary donation of SoHO to address unmet clinical needs, while contributing to training professionals and scientific literacy of our Society).
Collapse
Affiliation(s)
- Joaquín Delgadillo
- Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Transfusion Medicine Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Vantaa, Finland
| | - Allison Waters
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Emile van den Akker
- Department of Hematopoiesis and Sanquin Research, Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chantal Lechanteur
- University of Liège, Laboratory of Cell and Gene Therapy LTCG, Liège, Belgium
| | - Etienne Baudoux
- University of Liège, Laboratory of Cell and Gene Therapy LTCG, Liège, Belgium
| | - Nicola Gardiner
- Cryobiology Laboratory Stem Cell Facility, St. James's Hospital, Dublin, Ireland
| | - John De Vos
- Département d'ingénierie Cellulaire et Tissulaire, Unité de Thérapie Cellulaire, Hôpital Saint-Eloi, Montpellier, France
| | - Joaquim Vives
- Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Ding L, Oh S, Shrestha J, Lam A, Wang Y, Radfar P, Warkiani ME. Scaling up stem cell production: harnessing the potential of microfluidic devices. Biotechnol Adv 2023; 69:108271. [PMID: 37844769 DOI: 10.1016/j.biotechadv.2023.108271] [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: 04/29/2023] [Revised: 10/08/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Stem cells are specialised cells characterised by their unique ability to both self-renew and transform into a wide array of specialised cell types. The widespread interest in stem cells for regenerative medicine and cultivated meat has led to a significant demand for these cells in both research and practical applications. Despite the growing need for stem cell manufacturing, the industry faces significant obstacles, including high costs for equipment and maintenance, complicated operation, and low product quality and yield. Microfluidic technology presents a promising solution to the abovementioned challenges. As an innovative approach for manipulating liquids and cells within microchannels, microfluidics offers a plethora of advantages at an industrial scale. These benefits encompass low setup costs, ease of operation and multiplexing, minimal energy consumption, and the added advantage of being labour-free. This review presents a thorough examination of the prominent microfluidic technologies employed in stem cell research and explores their promising applications in the burgeoning stem cell industry. It thoroughly examines how microfluidics can enhance cell harvesting from tissue samples, facilitate mixing and cryopreservation, streamline microcarrier production, and efficiently conduct cell separation, purification, washing, and final cell formulation post-culture.
Collapse
Affiliation(s)
- Lin Ding
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia.
| | - Steve Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Jesus Shrestha
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Alan Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), Singapore, 138668, Singapore
| | - Yaqing Wang
- School of Biomedical Engineering, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Payar Radfar
- Smart MCs Pty Ltd, Ultimo, Sydney, 2007, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, NSW 2007, Australia..
| |
Collapse
|
44
|
Deszcz I. Stem Cell-Based Therapy and Cell-Free Therapy as an Alternative Approach for Cardiac Regeneration. Stem Cells Int 2023; 2023:2729377. [PMID: 37954462 PMCID: PMC10635745 DOI: 10.1155/2023/2729377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
The World Health Organization reports that cardiovascular diseases (CVDs) represent 32% of all global deaths. The ineffectiveness of conventional therapies in CVDs encourages the development of novel, minimally invasive therapeutic strategies for the healing and regeneration of damaged tissue. The self-renewal capacity, multilineage differentiation, lack of immunogenicity, and immunosuppressive properties of mesenchymal stem cells (MSCs) make them a promising option for CVDs. However, growing evidence suggests that myocardial regeneration occurs through paracrine factors and extracellular vesicle (EV) secretion, rather than through differentiation into cardiomyocytes. Research shows that stem cells secrete or surface-shed into their culture media various cytokines, chemokines, growth factors, anti-inflammatory factors, and EVs, which constitute an MSC-conditioned medium (MSC-CM) or the secretome. The use of MSC-CM enhances cardiac repair through resident heart cell differentiation, proliferation, scar mass reduction, a decrease in infarct wall thickness, and cardiac function improvement comparable to MSCs without their side effects. This review highlights the limitations and benefits of therapies based on stem cells and their secretome as an innovative treatment of CVDs.
Collapse
Affiliation(s)
- Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| |
Collapse
|
45
|
Giovannelli L, Bari E, Jommi C, Tartara F, Armocida D, Garbossa D, Cofano F, Torre ML, Segale L. Mesenchymal stem cell secretome and extracellular vesicles for neurodegenerative diseases: Risk-benefit profile and next steps for the market access. Bioact Mater 2023; 29:16-35. [PMID: 37456581 PMCID: PMC10338239 DOI: 10.1016/j.bioactmat.2023.06.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/01/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Neurodegenerative diseases represent a growing burden on healthcare systems worldwide. Mesenchymal stem cells (MSCs) have shown promise as a potential therapy due to their neuroregenerative, neuroprotective, and immunomodulatory properties, which are, however, linked to the bioactive substances they release, collectively known as secretome. This paper provides an overview of the most recent research on the safety and efficacy of MSC-derived secretome and extracellular vesicles (EVs) in clinical (if available) and preclinical models of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, Huntington's disease, acute ischemic stroke, and spinal cord injury. The article explores the biologically active substances within MSC-secretome/EVs, the mechanisms responsible for the observed therapeutic effects, and the strategies that may be used to optimize MSC-secretome/EVs production based on specific therapeutic needs. The review concludes with a critical discussion of current clinical trials and a perspective on potential future directions in translating MSC-secretome and EVs into the clinic, specifically regarding how to address the challenges associated with their pharmaceutical manufacturing, including scalability, batch-to-batch consistency, adherence to Good Manufacturing Practices (GMP) guidelines, formulation, and storage, along with quality controls, access to the market and relative costs, value for money and impact on total expenditure.
Collapse
Affiliation(s)
- Lorella Giovannelli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100, Novara, Italy
| | - Elia Bari
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100, Novara, Italy
| | - Claudio Jommi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100, Novara, Italy
| | | | - Daniele Armocida
- A.U.O, Policlinico Umberto I, Neurosurgery Division, Human Neurosciences Department, Sapienza University, 00135, Roma, Italy
| | - Diego Garbossa
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10126, Turin, Italy
| | - Fabio Cofano
- Department of Neuroscience Rita Levi Montalcini, Neurosurgery Unit, University of Turin, 10126, Turin, Italy
| | - Maria Luisa Torre
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100, Novara, Italy
- PharmaExceed S.r.l, 27100, Pavia, Italy
| | - Lorena Segale
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100, Novara, Italy
| |
Collapse
|
46
|
Blitzer GC, Glazer T, Burr A, Gustafson S, Ganz O, Meyers R, McDowell KA, Nickel KP, Mattison RJ, Weiss M, Chappell R, Rogus-Pulia NM, Galipeau J, Kimple RJ. Marrow-Derived Autologous Stromal Cells for the Restoration of Salivary Hypofunction (MARSH): A pilot, first-in-human study of interferon gamma-stimulated marrow mesenchymal stromal cells for treatment of radiation-induced xerostomia. Cytotherapy 2023; 25:1139-1144. [PMID: 37589639 PMCID: PMC10615723 DOI: 10.1016/j.jcyt.2023.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND AIMS Xerostomia, or the feeling of dry mouth, is a significant side effect of radiation therapy for patients with head and neck cancer (HNC). Preliminary data suggest that mesenchymal stromal/stem cells (MSCs) can improve salivary function. We performed a first-in-human pilot study of interferon gamma (IFNγ)-stimulated autologous bone marrow-derived MSCs, or MSC(M), for the treatment of radiation-induced xerostomia (RIX). Here we present the primary safety and secondary efficacy endpoints. METHODS A single-center pilot clinical trial was conducted investigating the safety and tolerability of autologous IFNγ-stimulated MSC(M). The study was conducted under an approved Food and Drug Administration Investigational New Drug application using an institutional review board-approved protocol (NCT04489732). Patients underwent iliac crest bone marrow aspirate and MSC(M) were isolated, cultured, stimulated with IFNγ and cryopreserved for later use. Banked cells were thawed and allowed to recover in culture before patients received a single injection of 10 × 106 MSC(M) into the right submandibular gland under ultrasound guidance. The primary objective was determination of safety and tolerability by evaluating dose-limiting toxicity (DLT). A DLT was defined as submandibular pain >5 on a standard 10-point pain scale or any serious adverse event (SAE) within 1 month after injection. Secondary objectives included analysis of efficacy as measured by salivary quantification and using three validated quality of life instruments. Quantitative results are reported as mean and standard deviation. RESULTS Six patients with radiation-induced xerostomia who had completed radiation at least 2 years previously (average 7.8 years previously) were enrolled in the pilot study. The median age was 71 (61-74) years. Five (83%) patients were male. Five patients (83%) were treated with chemoradiation and one patient (17%) with radiation alone. Grade 1 pain was seen in 50% of patients after submandibular gland injection; all pain resolved within 4 days. No patients reported pain 1 month after injection, with no SAE or other DLTs reported 1 month after injection. The analysis of secondary endpoints demonstrated a trend of increased salivary production. Three patients (50%) had an increase in unstimulated saliva at 1 and 3 months after MSC(M) injection. Quality of life surveys also showed a trend toward improvement. CONCLUSIONS Injection of autologous IFNγ-stimulated MSC(M) into a singular submandibular gland of patients with RIX is safe and well tolerated in this pilot study. A trend toward an improvement in secondary endpoints of salivary quantity and quality of life was observed. This first-in-human study provides support for further investigation into IFNγ-stimulated MSC(M) injected in both submandibular glands as an innovative approach to treat RIX and improve quality of life for patients with HNC.
Collapse
Affiliation(s)
- Grace C Blitzer
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| | - Tiffany Glazer
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Adam Burr
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Sara Gustafson
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Olga Ganz
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ross Meyers
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kimberly A McDowell
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Kwangok P Nickel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ryan J Mattison
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Marissa Weiss
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Richard Chappell
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Nicole M Rogus-Pulia
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jacques Galipeau
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Randall J Kimple
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA; UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| |
Collapse
|
47
|
Smolinská V, Boháč M, Danišovič Ľ. Current status of the applications of conditioned media derived from mesenchymal stem cells for regenerative medicine. Physiol Res 2023; 72:S233-S245. [PMID: 37888967 PMCID: PMC10669946 DOI: 10.33549/physiolres.935186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 12/01/2023] Open
Abstract
Recently published studies suggest that the paracrine substances released by mesenchymal stem cells (MSCs) are the primary motive behind the therapeutic action reported in these cells. Pre-clinical and clinical research on MSCs has produced promising outcomes. Furthermore, these cells are generally safe for therapeutic use and may be extracted from a variety of anatomical regions. Recent research has indicated, however, that transplanted cells do not live long and that the advantages of MSC treatment may be attributable to the large diversity of bioactive substances they create, which play a crucial role in the control of essential physiological processes. Secretome derivatives, such as conditioned media or exosomes, may provide significant benefits over cells in terms of manufacture, preservation, handling, longevity of the product, and potential as a ready-to-use biologic product. Despite their immunophenotypic similarities, the secretome of MSCs appears to vary greatly depending on the host's age and the niches in which the cells live. The secretome's effect on multiple biological processes such as angiogenesis, neurogenesis, tissue repair, immunomodulation, wound healing, anti-fibrotic, and anti-tumor for tissue maintenance and regeneration has been discovered. Defining the secretome of cultured cultivated MSC populations by conditioned media analysis will allow us to assess its potential as a novel treatment approach. This review will concentrate on accumulating data from pre-clinical and clinical trials pointing to the therapeutic value of the conditioned medium. At last, the necessity of characterizing the conditioned medium for determining its potential for cell-free treatment therapy will be emphasized in this study.
Collapse
|
48
|
Haddouti EM, Reinhardt N, Ossendorff R, Burger C, Wirtz DC, de la Fuente M, Schildberg FA. Effects of single and repeated shock wave application on the osteogenic differentiation potential of human primary mesenchymal stromal cells and the osteoblastic cell line MG63 in vitro. Front Bioeng Biotechnol 2023; 11:1207655. [PMID: 37901841 PMCID: PMC10602737 DOI: 10.3389/fbioe.2023.1207655] [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: 04/17/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction: Extracorporeal shock wave therapy is a non-invasive and effective option for treating various musculoskeletal disorders. Recent literature indicates that the parameters for extracorporeal shock wave therapy, such as the optimal intensity, treatment frequency, and localization, are yet to be determined. Studies reporting on the effects of shock wave application on primary mesenchymal stromal cells (MSCs) as well as osteoblastic cell lines in vitro are barely available and not standardized. Methods: In this study, we designed a special setup to precisely expose primary MSCs and the osteoblastic cell line MG63 to shock waves and subsequently analyzed the resulting cellular responses using standardized protocols to investigate their viability, proliferation behavior, cytokine secretion, and osteogenic differentiation potential in vitro. The shock wave transducer was coupled to a specifically designed water bath containing a 5 mL tube holder. Primary human MSCs and MG63 cells were trypsinated and centrifuged in a 5 mL tube and exposed to single and repeated shock wave application using different intensities and numbers of pulses. Results: Single treatment of MSCs using intensities 5, 10, 15, and 20 and pulse numbers 100, 250, 500, 750, and 1,000 at a constant pulse repetition frequency of 1 Hz resulted in a decreased viability and proliferation of both cell types with an increase in the intensity and number of pulses compared to controls. No significant difference in the osteogenic differentiation was observed at different time intervals in both cell types when a single shock wave application was performed. However, repeated shock wave sessions over three consecutive days of primary MSCs using low intensity levels 0.1 and 1 showed significant osteogenic differentiation 4-fold higher than that of the extracted Alizarin Red S at day 14, whereas MG63 cells showed no significant osteogenic differentiation compared to their corresponding controls. More specifically, repeated shock wave application triggered a significant downregulation of COL1A1, upregulation of RUNX2, and sustained increase of OCN in primary MSCs but not in the cell line MG63 when induced toward the osteogenic differentiation. Discussion: The effects of shock wave application on MSCs make it an effective therapy in regenerative medicine. We established a protocol to analyze a standardized shock wave application on MSCs and were able to determine conditions that enhance the osteogenic differentiation of MSCs in vitro.
Collapse
Affiliation(s)
- El-Mustapha Haddouti
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Nina Reinhardt
- Chair of Medical Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Matias de la Fuente
- Chair of Medical Engineering, Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
49
|
Mora-Boza A, Ghebrezadik SG, Leisen JE, García AJ. Rapid and Facile Light-Based Approach to Fabricate Protease-Degradable Poly(ethylene glycol)-norbornene Microgels for Cell Encapsulation. Adv Healthc Mater 2023; 12:e2300942. [PMID: 37235850 PMCID: PMC10592588 DOI: 10.1002/adhm.202300942] [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: 03/23/2023] [Revised: 05/15/2023] [Indexed: 05/28/2023]
Abstract
Thiol-norbornene photoclickable poly (ethylene glycol) (PEG)-based (PEG-NB) hydrogels are attractive biomaterials for cell encapsulation, drug delivery, and regenerative medicine applications. Although many crosslinking strategies and chemistries have been developed for PEG-NB bulk hydrogels, fabrication approaches of PEG-NB microgels have not been extensively explored. Here, a fabrication strategy for 4-arm amide-linked PEG-NB (PEG-4aNB) microgels using flow-focusing microfluidics for human mesenchymal stem/stromal cell (hMSCs) encapsulation is presented. PEG-4aNB photochemistry allows high-throughput, ultrafast generation, and cost-effective synthesis of monodispersed microgels (diameter 340 ± 18, 380 ± 24, and 420 ± 15 µm, for 6, 8, and 10 wt% of PEG-4aNB, respectively) using an in situ crosslinking methodology in a microfluidic device. PEG-4aNB microgels show in vitro degradability due to the incorporation of a protease-degradable peptide during photocrosslinking and encapsulated cells show excellent viability and metabolic activity for at least 13 days of culture. Furthermore, the secretory profile (i.e., MMP-13, ICAM-1, PD-L1, CXCL9, CCL3/MIP-1, IL-6, IL-12, IL-17E, TNF-α, CCL2/MCP-1) of encapsulated hMSCs shows increased expression in response to IFN-γ stimulation. Collectively, this work shows a versatile and facile approach for the fabrication of protease-degradable PEG-4aNB microgels for cell encapsulation.
Collapse
Affiliation(s)
- Ana Mora-Boza
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- CÚRAM, University of Galway, Galway, H91 W2TY, Ireland
| | - Saron G Ghebrezadik
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Agnes Scott College, Decatur, GA, 30030, USA
| | - Johannes E Leisen
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332-0363, USA
| |
Collapse
|
50
|
Torrents S, del Moral AE, Codinach M, Rodríguez L, Querol S, Vives J. Optimized reagents for immunopotency assays on mesenchymal stromal cells for clinical use. Immunol Res 2023; 71:725-734. [PMID: 37120479 PMCID: PMC10148700 DOI: 10.1007/s12026-023-09385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023]
Abstract
Multipotent mesenchymal stromal cells (MSC) offer new therapeutic opportunities based on their ability to modulate an imbalanced immune system. Immunomodulatory potency is typically demonstrated in vitro by measuring the presence of surrogate markers (i.e., indoleamine-2,3-dioxygenase, IDO; tumor necrosis factor receptor type 1, TNFR1) and/or functional assays in co-cultures (i.e., inhibition of lymphoproliferation, polarization of macrophages). However, the biological variability of reagents used in the latter type of assays leads to unreliable and difficult to reproduce data therefore making cross-comparison between batches difficult, both at the intra- and inter-laboratory levels. Herein, we describe a set of experiments aiming at the definition and validation of reliable biological reagents as a first step towards standardization of a potency assay. This approach is based on the co-culture of Wharton's jelly (WJ)-derived MSC and cryopreserved pooled peripheral blood mononuclear cells. Altogether, we successfully defined a robust and reproducible immunopotency assay based on previously described methods incorporating substantial improvements such as cryopreservation of multiple vials of pooled peripheral blood mononuclear cells (PBMC) from 5 individual donors that enable a number of tests with same reagents, also reducing waste of PBMC from individual donors and therefore contributing to a more efficient and ethical method to use substances of human origin (SoHO). The new methodology was successfully validated using 11 batches of clinical grade MSC,WJ. Methods described here contribute to minimize PBMC donor variability while reducing costs, streamlining assay setup and convenience and laying the foundations for harmonization of biological reagents usage in standardized immunopotency assays for MSC. HIGHLIGHTS: • The use of pools of peripheral blood mononuclear cells (PBMCs) in potency assays contributes to robust and reproducible results, which is key in the assessment of mesenchymal stroma cells (MSC) potency for batch release. • Cryopreservation of PBMCs does not impact negatively on their activation and proliferation abilities. • Cryopreserved pools of PBMC constitutes convenient off-the-shelf reagents for potency assays. • Cryopreservation of pooled PBMCs from multiple donors is a way to reduce waste of donated PBMC and its associated costs, as well as reducing the impact of individual donor variability of substances of human origin (SoHO).
Collapse
Affiliation(s)
- Sílvia Torrents
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
- Transfusion Medicine Group, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
| | - Andrés Escudero del Moral
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Margarita Codinach
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
- Transfusion Medicine Group, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
| | - Luciano Rodríguez
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
- Transfusion Medicine Group, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
| | - Sergi Querol
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
- Transfusion Medicine Group, Vall d’Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
| | - Joaquim Vives
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
- Musculoskeletal Tissue Engineering Group, Vall d’Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
- Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de La Vall d’Hebron 129-139, 08035 Barcelona, Spain
| |
Collapse
|