1
|
Hu Y, Zheng L, Zheng Z, Fu M, Peng H, Ma S. Microbead Encapsulation Strategy for Efficient Production of Extracellular Vesicles Derived From Human Mesenchymal Stem Cells. J Extracell Vesicles 2025; 14:e70053. [PMID: 40240906 PMCID: PMC12003097 DOI: 10.1002/jev2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/07/2025] [Indexed: 04/18/2025] Open
Abstract
Human mesenchymal stem cell-derived extracellular vesicles (hMSC-EVs) have shown great potential in tissue repair and regeneration. However, their scalable production and functional quality are still limited by current expansion technologies. In this study, we propose a production technology for hMSC-EVs based on three-dimensional (3D) microbead culture, which enhances the secretory behaviour of hMSC. Fixed number of MSCs were encapsulated in Matrigel at appropriate densities and printed into 3D microbeads by the custom automated microfluidic bead-jet printing technique. Compared with 2D culture group, EVs derived from 3D hMSC microbead had smaller size and increased yield by 20-fold, and the actin depolymerisation of the cell may be an important mechanism for enhancing EV secretion. Further analysis confirmed that the EVs derived from 3D hMSC microbead exhibited enhanced angiogenic and proliferative capabilities, which promoted the viability and tube-forming capacity of human umbilical vein endothelial cells (HUVEC). In conclusion, this automated microfluidic microbead encapsulation technology increased the yield and therapeutic effect of hMSC-EVs and provides a platform for scalable EV production of regenerative therapies.
Collapse
Affiliation(s)
- Yunxia Hu
- Institute of Biopharmaceutical and Health Engineering (iBHE), Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
- Key Lab of Industrial BiocatalysisMinistry of EducationShenzhenChina
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education InstitutesTsinghua Shenzhen International Graduate SchoolShenzhenChina
| | - Lijuan Zheng
- Institute of Biopharmaceutical and Health Engineering (iBHE), Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
- Key Lab of Industrial BiocatalysisMinistry of EducationShenzhenChina
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education InstitutesTsinghua Shenzhen International Graduate SchoolShenzhenChina
| | - Zheng Zheng
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Mali Fu
- Shenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Haiying Peng
- General Hospital of the Southern Theater Command of the Chinese People's Liberation ArmyGuangzhouChina
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering (iBHE), Tsinghua Shenzhen International Graduate School (SIGS)Tsinghua UniversityShenzhenChina
- Key Lab of Industrial BiocatalysisMinistry of EducationShenzhenChina
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education InstitutesTsinghua Shenzhen International Graduate SchoolShenzhenChina
| |
Collapse
|
2
|
Jiu J, Liu H, Li D, Li X, Zhang J, Yan L, Fan Z, Li S, Du G, Li JJ, Wu A, Liu W, Du Y, Zhao B, Wang B. 3D Mechanical Response Stem Cell Complex Repairs Spinal Cord Injury by Promoting Neurogenesis and Regulating Tissue Homeostasis. Adv Healthc Mater 2025; 14:e2404925. [PMID: 39853962 DOI: 10.1002/adhm.202404925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Indexed: 01/26/2025]
Abstract
Spinal cord injury (SCI) leads to acute tissue damage that disrupts the microenvironmental homeostasis of the spinal cord, inhibiting cell survival and function, and thereby undermining treatment efficacy. Traditional stem cell therapies have limited success in SCI, due to the difficulties in maintaining cell survival and inducing sustained differentiation into neural lineages. A new solution may arise from controlling the fate of stem cells by creating an appropriate mechanical microenvironment. In this study, mechanical response stem cell complex (MRSCC) is created as an innovative therapeutic strategy for SCI, utilizing 3D bioprinting technology and gelatin microcarriers (GM) loaded with mesenchymal stem cells (MSCs). GM creates an optimal microenvironment for MSCs growth and paracrine activity. Meanwhile, 3D bioprinting allows accurate control of spatial pore architecture and mechanical characteristics of the cell construct to encourage neuroregeneration. The mechanical microenvironment created by MRSCC is found to activate the Piezo1 channel and prevent excessive nuclear translocation of YAP, thereby increasing neural-related gene expression in MSCs. Transplanting MRSCC in rats with spinal cord injuries boosts sensory and motor recovery, reduces inflammation, and stimulates the regeneration of neurons and glial cells. The MRSCC offers a new tissue engineering solution that can promote spinal cord repair.
Collapse
Affiliation(s)
- Jingwei Jiu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Haifeng Liu
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Dijun Li
- Department of Orthopedics, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443000, China
| | - Xiaoke Li
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Zhang
- Department of Emergency Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, China
| | - Lei Yan
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zijuan Fan
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Songyan Li
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Guangyuan Du
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, Sydney, NSW, 2007, Australia
| | - Aimin Wu
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wei Liu
- Development of Research, Beijing Hua Niche Biotechnology Co., LTD, Beijing, 100084, China
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bin Zhao
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Bin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| |
Collapse
|
3
|
Wang Q, Guo W, Niu L, Zhou Y, Wang Z, Chen J, Chen J, Ma J, Zhang J, Jiang Z, Wang B, Zhang Z, Li C, Jian Z. 3D-hUMSCs Exosomes Ameliorate Vitiligo by Simultaneously Potentiating Treg Cells-Mediated Immunosuppression and Suppressing Oxidative Stress-Induced Melanocyte Damage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404064. [PMID: 38887870 PMCID: PMC11336971 DOI: 10.1002/advs.202404064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/15/2024] [Indexed: 06/20/2024]
Abstract
Vitiligo is an autoimmune disease characterized by epidermal melanocyte destruction, with abnormal autoimmune responses and excessive oxidative stress as two cardinal mechanisms. Human umbilical mesenchymal stem cells-derived exosomes (hUMSCs-Exos) are regarded as promising therapeutic choice for autoimmune diseases due to potent immunosuppressive and anti-oxidative properties, which can be potentiated under 3D cell culture condition. Nevertheless, whether exosomes derived from 3D spheroids of hUMSCs (3D-Exos) exhibit considerable therapeutic effect on vitiligo and the underlying mechanism remain elusive. In this study, systemic administration of 3D-Exos showed a remarkable effect in treating mice with vitiligo, as revealed by ameliorated skin depigmentation, less CD8+T cells infiltration, and expanded Treg cells in skin, and 3D-Exos exerted a better effect than 2D-Exos. Mechanistically, 3D-Exos can prominently facilitate the expansion of Treg cells in vitiligo lesion and suppress H2O2-induced melanocytes apoptosis. Forward miRNA profile analysis and molecular experiments have demonstrated that miR-132-3p and miR-125b-5p enriched in 3D-Exos greatly contributed to these biological effects by targeting Sirt1 and Bak1 respectively. In aggregate, 3D-Exos can efficiently ameliorate vitiligo by simultaneously potentiating Treg cells-mediated immunosuppression and suppressing oxidative stress-induced melanocyte damage via the delivery of miR-132-3p and miR-125b-5p. The employment of 3D-Exos will be a promising treament for vitiligo.
Collapse
Affiliation(s)
- Qi Wang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Weinan Guo
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Liaoran Niu
- Department of Digestive SurgeryXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Yuqi Zhou
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Zeqian Wang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jianru Chen
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jiaxi Chen
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jingjing Ma
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jia Zhang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Zhaoting Jiang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Bo Wang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Zhe Zhang
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Chunying Li
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Zhe Jian
- Department of DermatologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| |
Collapse
|
4
|
Castilla-Casadiego DA, Morton LD, Loh DH, Pineda-Hernandez A, Chavda AP, Garcia F, Rosales AM. Peptoid-Cross-Linked Hydrogel Stiffness Modulates Human Mesenchymal Stromal Cell Immunoregulatory Potential in the Presence of Interferon-Gamma. Macromol Biosci 2024; 24:e2400111. [PMID: 38567626 PMCID: PMC11250919 DOI: 10.1002/mabi.202400111] [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: 03/22/2024] [Indexed: 04/04/2024]
Abstract
Human mesenchymal stromal cell (hMSC) manufacturing requires the production of large numbers of therapeutically potent cells. Licensing with soluble cytokines improves hMSC therapeutic potency by enhancing secretion of immunoactive factors but typically decreases proliferative ability. Soft hydrogels, however, have shown promise for boosting immunomodulatory potential, which may compensate for decreased proliferation. Here, hydrogels are cross-linked with peptoids of different secondary structures to generate substrates of various bulk stiffnesses but fixed network connectivity. Secretions of interleukin 6, monocyte chemoattractive protein-1, macrophage colony-stimulating factor, and vascular endothelial growth factor are shown to depend on hydrogel stiffness in the presence of interferon gamma (IFN-γ) supplementation, with soft substrates further improving secretion. The immunological function of these secreted cytokines is then investigated via coculture of hMSCs seeded on hydrogels with primary peripheral blood mononuclear cells (PBMCs) in the presence and absence of IFN-γ. Cocultures with hMSCs seeded on softer hydrogels show decreased PBMC proliferation with IFN-γ. To probe possible signaling pathways, immunofluorescent studies probe the nuclear factor kappa B pathway and demonstrate that IFN-γ supplementation and softer hydrogel mechanics lead to higher activation of this pathway. Overall, these studies may allow for production of more efficacious therapeutic hMSCs in the presence of IFN-γ.
Collapse
Affiliation(s)
| | - Logan D. Morton
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Darren H. Loh
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Aldaly Pineda-Hernandez
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ajay P. Chavda
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Francis Garcia
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| | - Adrianne M. Rosales
- Mcketta Department of Chemical Engineering, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
5
|
Gao M, Guo H, Dong X, Wang Z, Yang Z, Shang Q, Wang Q. Regulation of inflammation during wound healing: the function of mesenchymal stem cells and strategies for therapeutic enhancement. Front Pharmacol 2024; 15:1345779. [PMID: 38425646 PMCID: PMC10901993 DOI: 10.3389/fphar.2024.1345779] [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: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
A wound takes a long time to heal and involves several steps. Following tissue injury, inflammation is the primary cause of tissue regeneration and repair processes. As a result, the pathophysiological processes involving skin damage, healing, and remodeling depend critically on the control of inflammation. The fact that it is a feasible target for improving the prognosis of wound healing has lately become clear. Mesenchymal stem cells (MSCs) are an innovative and effective therapeutic option for wound healing due to their immunomodulatory and paracrine properties. By controlling the inflammatory milieu of wounds through immunomodulation, transplanted MSCs have been shown to speed up the healing process. In addition to other immunomodulatory mechanisms, including handling neutrophil activity and modifying macrophage polarization, there may be modifications to the activation of T cells, natural killer (NK) cells, and dendritic cells (DCs). Furthermore, several studies have shown that pretreating MSCs improves their ability to modulate immunity. In this review, we summarize the existing knowledge about how MSCs influence local inflammation in wounds by influencing immunity to facilitate the healing process. We also provide an overview of MSCs optimizing techniques when used to treat wounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qiying Wang
- Department of Plastic Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Tan Y, Ding Y, Liu S, Liu P, Cai K. Titanium-based substrate modified with nanoenzyme for accelerating the repair of bone defect. Colloids Surf B Biointerfaces 2024; 234:113737. [PMID: 38176336 DOI: 10.1016/j.colsurfb.2023.113737] [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] [Revised: 12/24/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Titanium (Ti) and titanium alloy are the most common metal materials in clinical orthopedic surgery. However, in the initial stage of surgery and implantation, the production of excessive reactive oxygen species (ROS) can induce oxidative stress (OS) microenvironment. OS will further inhibit the growth of new bone, resulting in surgical failure. In this study, based on the fact that nanoscale manganese dioxide (MnO2) can show H2O2-like enzyme activity, a MnO2 nanocoating was prepared on mciro-nano structured surface of Ti substrate via a two-step method of alkaline thermal and hydrothermal treatment. The results of scanning electron microscopy (SEM), X-ray diffractometer (XRD) and X-ray photoelectron spectroscopy (XPS) showed that the nano-MnO2 coating was successfully fabricated on the surface of Ti substrate. The results of measurement of H2O2, dissolved O2 and intracellular ROS in vitro showed that the treated Ti substrate could efficiently eliminate H2O2 and reduce ROS. Furthermore, the modified Ti substrate could promote the early adhesion, proliferation and osteogenic differentiation of MSCs, which was demonstrated by experimental results of cell morphology, cell viability, alkaline phosphatase, collagen, and mineralization deposition. The results of quantitative real-time polymerase chain reaction (qRT-PCR) of MSCs adhered the modified Ti substrate showed that the expression of genes related to osteogenic differentiation significantly increased. More importantly, the modified Ti implant could eliminate ROS at the injury site, reduce OS and promote the regeneration of bone tissue, which was demonstrated via hematoxylin/eosin, Masson's trichrome and immunohistochemical staining. In conclusion, the modified Ti implant presented here had the effect of reducing OS and promoting osseointegration. Relevant research ideas and results provide new methods for the research and development of functional implants, which have potential application value in the field of orthopedics.
Collapse
Affiliation(s)
- Yingying Tan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing China
| | - Yao Ding
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing China
| | - Shaopeng Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing China
| | - Peng Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing China.
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing China.
| |
Collapse
|
7
|
Li J, Zhang J, Ye H, Wang Q, Ouyang Y, Luo Y, Gong Y. Pulmonary decellularized extracellular matrix (dECM) modified polyethylene terephthalate three-dimensional cell carriers regulate the proliferation and paracrine activity of mesenchymal stem cells. Front Bioeng Biotechnol 2024; 11:1324424. [PMID: 38260733 PMCID: PMC10800494 DOI: 10.3389/fbioe.2023.1324424] [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: 10/19/2023] [Accepted: 12/07/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction: Mesenchymal stem cells (MSCs) possess a high degree of self-renewal capacity and in vitro multi-lineage differentiation potential. Decellularized materials have garnered considerable attention due to their elevated biocompatibility, reduced immunogenicity, excellent biodegradability, and the ability to partially mimic the in vivo microenvironment conducive to cell growth. To address the issue of mesenchymal stem cells losing their stem cell characteristics during two-dimensional (2D) cultivation, this study established three-dimensional cell carriers modified with lung decellularized extracellular matrix and assessed its impact on the life activities of mesenchymal stem cells. Methods: This study employed PET as a substrate material, grafting with polydopamine (PDA), and constructing a decellularized extracellular matrix (dECM) coating on its surface, thus creating the PET/PDA/dECM three-dimensional (3D) composite carrier. Subsequently, material characterization of the cellular carriers was conducted, followed by co-culturing with human umbilical cord mesenchymal stem cells in vitro, aiming to investigate the material's impact on the proliferation and paracrine activity of mesenchymal stem cells. Results and Discussion: Material characterization demonstrated successful grafting of PDA and dECM materials, and it had complete hydrophilicity, high porosity, and excellent mechanical properties. The material was rich in various ECM proteins (collagen I, collagen IV , laminin, fibronectin, elastin), indicating good biocompatibility. In long-term in vitro cultivation (14 days) experiments, the PET/PDA/dECM three-dimensional composite carrier significantly enhanced adhesion and proliferation of human umbilical cord-derived mesenchymal stem cells (HUCMSCs), with a proliferation rate 1.9 times higher than that of cells cultured on tissue culture polystyrene (TCPS) at day 14. Furthermore, it effectively maintained the stem cell characteristics, expressing specific antigens for HUCMSCs. Through qPCR, Western blot, and ELISA experiments, the composite carrier markedly promoted the expression and secretion of key cell factors in HUCMSCs. These results demonstrate that the PET/PDA/dECM composite carrier holds great potential for scaling up MSCs' long-term in vitro cultivation and the production of paracrine factors.
Collapse
Affiliation(s)
- Jinze Li
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Jiali Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Hao Ye
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Qixuan Wang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Yanran Ouyang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
| | - Yuxi Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, Sun Yat-Sen University, Guangzhou, China
| | - Yihong Gong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, China
- Guangdong Provincial Key Laboratory of Sensor Technology and Biomedical Instrument, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
8
|
Mahmoud M, Abdel-Rasheed M, Galal ER, El-Awady RR. Factors Defining Human Adipose Stem/Stromal Cell Immunomodulation in Vitro. Stem Cell Rev Rep 2024; 20:175-205. [PMID: 37962697 PMCID: PMC10799834 DOI: 10.1007/s12015-023-10654-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Human adipose tissue-derived stem/stromal cells (hASCs) are adult multipotent mesenchymal stem/stromal cells with immunomodulatory capacities. Here, we present up-to-date knowledge on the impact of different experimental and donor-related factors on hASC immunoregulatory functions in vitro. The experimental determinants include the immunological status of hASCs relative to target immune cells, contact vs. contactless interaction, and oxygen tension. Factors such as the ratio of hASCs to immune cells, the cellular context, the immune cell activation status, and coculture duration are also discussed. Conditioning of hASCs with different approaches before interaction with immune cells, hASC culture in xenogenic or xenofree culture medium, hASC culture in two-dimension vs. three-dimension with biomaterials, and the hASC passage number are among the experimental parameters that greatly may impact the hASC immunosuppressive potential in vitro, thus, they are also considered. Moreover, the influence of donor-related characteristics such as age, sex, and health status on hASC immunomodulation in vitro is reviewed. By analysis of the literature studies, most of the indicated determinants have been investigated in broad non-standardized ranges, so the results are not univocal. Clear conclusions cannot be drawn for the fine-tuned scenarios of many important factors to set a standard hASC immunopotency assay. Such variability needs to be carefully considered in further standardized research. Importantly, field experts' opinions may help to make it clearer.
Collapse
Affiliation(s)
- Marwa Mahmoud
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt.
- Department of Medical Molecular Genetics, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt.
| | - Mazen Abdel-Rasheed
- Stem Cell Research Group, Medical Research Centre of Excellence, National Research Centre, 33 El Buhouth St, Ad Doqi, Dokki, 12622, Cairo Governorate, Egypt
- Department of Reproductive Health Research, National Research Centre, Cairo, Egypt
| | - Eman Reda Galal
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Rehab R El-Awady
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
9
|
Dunn CM, Kameishi S, Parker T, Cho YK, Song SU, Grainger DW, Okano T. Cellular Interactions in Cell Sheets Enhance Mesenchymal Stromal Cell Immunomodulatory Properties. Tissue Eng Part A 2023; 29:594-603. [PMID: 37847176 DOI: 10.1089/ten.tea.2023.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023] Open
Abstract
Immune-related applications of mesenchymal stromal cells (MSCs) in cell therapy seek to exploit immunomodulatory paracrine signaling pathways to reduce inflammation. A key MSC therapeutic challenge is reducing patient outcome variabilities attributed to insufficient engraftment/retention of injected heterogenous MSCs. To address this, we propose directly transplantable human single-cell-derived clonal bone marrow MSC (hcBMSC) sheets. Cell sheet technology is a scaffold-free tissue engineering strategy enabling scalable production of highly engraftable cell constructs retaining endogenous cell-cell and cell-matrix interactions, important to cell function. cBMSCs, as unique MSC subset populations, facilitate rational selection of therapeutically relevant MSC clones from donors. Here, we combine human cBMSCs with cell sheet technology, demonstrating cell sheet fabrication as a method to significantly upregulate expression of immunomodulatory molecules interleukin (IL)-10, indoleamine 2,3-dioxygenase (IDO-1), and prostaglandin E synthase 2 (PTGES2) across GMP-grade hcBMSC lines and whole human bone marrow-derived MSCs compared to respective conventional cell suspensions. When treated with carbenoxolone, a gap junction inhibitor, cell sheets downregulate IL-10 and IDO-1 expression, implicating functional roles for intercellular sheet interactions. Beyond producing directly transferable multicellular hcBMSC constructs, cell sheet technology amplifies hcBMSC expression of immunomodulatory factors important to therapeutic action. In addition, this work demonstrates the importance of cell-cell interactions as a tissue engineering design criterion to enhance consistent MSC functions.
Collapse
Affiliation(s)
- Celia M Dunn
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Sumako Kameishi
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
| | - Tavie Parker
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | | | - Sun U Song
- SCM Lifescience Co., Ltd., Incheon, Republic of Korea
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, Utah, USA
- Institute for Advanced Biomedical Sciences, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
10
|
Vimalraj S, Saravanan S. Tooth-derived stem cells integrated biomaterials for bone and dental tissue engineering. Cell Tissue Res 2023; 394:245-255. [PMID: 37548764 DOI: 10.1007/s00441-023-03815-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Recent years have seen the emergence of tissue engineering strategies as a means to overcome some of the limits of conventional medical treatment. A biomaterial with tailored physio-chemical characteristics is used in this sophisticated method to transport stem cells and growth factors/bioactive substances, or to attract local endogenous cells, enabling new tissue formation. Biomaterials might serve as a biomimetic structure inspired by the natural milieu, assisting the cells in establishing their natural relationships. Such a method would benefit from having ready access to an abundant reservoir of stem cells with strong tissue regeneration capacity, in addition to using biological compatible material to promote new tissue creation. Teeth may have a plethora of self-renewing, multipotent mesenchymal stem cell (MSC) populations. Recent advancements and promising directions for cell transplantation and homing techniques using dental MSCs for tissue regeneration are discussed in this review paper. Overall, this research paints a picture of the present landscape of new approaches to using tooth-derived MSCs in conjunction with biomaterials and bioactive substances for tissue regeneration.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, 600 077, Tamil Nadu, India.
| | - Sekaran Saravanan
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, 600 077, Tamil Nadu, India
| |
Collapse
|
11
|
Morán MDC, Cirisano F, Ferrari M. Spheroid Formation and Recovery Using Superhydrophobic Coating for Regenerative Purposes. Pharmaceutics 2023; 15:2226. [PMID: 37765195 PMCID: PMC10538210 DOI: 10.3390/pharmaceutics15092226] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Cell therapies commonly pursue tissue stimulation for regenerative purposes by replacing cell numbers or supplying for functional deficiencies. To this aim, monodispersed cells are usually transplanted for incorporation by local injection. The limitations of this strategy include poor success associated with cell death, insufficient retention, or cell damage due to shear forces associated with the injection. Spheroids have recently emerged as a model that mimics an in vivo environment with more representative cell-to-cell interactions and better intercellular communication. Nevertheless, cost-effective and lab friendly fabrication and effectively performed recovery are challenges that restrict the broad application of spheroids. In this work, glass surfaces were modified with an environmentally friendly superhydrophobic coating. The superhydrophobic surfaces were used for the 3D spheroid preparation of fibroblasts (3T3 cell line) and keratinocytes (HaCaT cell line). The effectiveness of the spheroids to be recovered and grown under 2D culture conditions was evaluated. The morphology of the migrated cells from the 3D spheroids was characterized at the nano-microscale through 3D profilometry. The results demonstrated improved adhesion and proliferation in the migrated cells, both advanced properties for regenerative applications.
Collapse
Affiliation(s)
- María del Carmen Morán
- Departament de Bioquímica i Fisiologia, Secció de Fisiologia—Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, Avda. Joan XXIII, 27-31, 08028 Barcelona, Spain
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
| | - Francesca Cirisano
- CNR-ICMATE Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia, Via De Marini, 6, 16149 Genova, Italy;
| | - Michele Ferrari
- Institut de Nanociència i Nanotecnologia—IN2UB, Universitat de Barcelona, Avda. Diagonal, 645, 08028 Barcelona, Spain
- CNR-ICMATE Istituto di Chimica della Materia Condensata e di Tecnologie per l’Energia, Via De Marini, 6, 16149 Genova, Italy;
| |
Collapse
|
12
|
Miceli V, Zito G, Bulati M, Gallo A, Busà R, Iannolo G, Conaldi PG. Different priming strategies improve distinct therapeutic capabilities of mesenchymal stromal/stem cells: Potential implications for their clinical use. World J Stem Cells 2023; 15:400-420. [PMID: 37342218 PMCID: PMC10277962 DOI: 10.4252/wjsc.v15.i5.400] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) have shown significant therapeutic potential, and have therefore been extensively investigated in preclinical studies of regenerative medicine. However, while MSCs have been shown to be safe as a cellular treatment, they have usually been therapeutically ineffective in human diseases. In fact, in many clinical trials it has been shown that MSCs have moderate or poor efficacy. This inefficacy appears to be ascribable primarily to the heterogeneity of MSCs. Recently, specific priming strategies have been used to improve the therapeutic properties of MSCs. In this review, we explore the literature on the principal priming approaches used to enhance the preclinical inefficacy of MSCs. We found that different priming strategies have been used to direct the therapeutic effects of MSCs toward specific pathological processes. Particularly, while hypoxic priming can be used primarily for the treatment of acute diseases, inflammatory cytokines can be used mainly to prime MSCs in order to treat chronic immune-related disorders. The shift in approach from regeneration to inflammation implies, in MSCs, a shift in the production of functional factors that stimulate regenerative or anti-inflammatory pathways. The opportunity to fine-tune the therapeutic properties of MSCs through different priming strategies could conceivably pave the way for optimizing their therapeutic potential.
Collapse
Affiliation(s)
- Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy.
| | - Giovanni Zito
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Matteo Bulati
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Rosalia Busà
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Pier Giulio Conaldi
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| |
Collapse
|
13
|
Li X, Li X, Yang J, Lin J, Zhu Y, Xu X, Cui W. Living and Injectable Porous Hydrogel Microsphere with Paracrine Activity for Cartilage Regeneration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207211. [PMID: 36651038 DOI: 10.1002/smll.202207211] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
Paracrine is an important mechanism in mesenchymal stem cells (MSCs) that promotes tissue regeneration. However, anoikis is attributed to unsuitable adhesion microenvironment hindered this paracrine effect. In this study, a living and injectable porous hydrogel microsphere with long-term paracrine activity is constructed via the freeze-drying microfluidic technology and the incorporation of platelet-derived growth factor-BB (PDGF-BB) and exogenous MSCs. Benefiting from the porous structure and superior mechanical property of methacrylate gelatin (GelMA) hydrogel microspheres (GMs), exogenous stem cells are able to adhere and proliferate on GMs, thereby facilitating cell-to-extracellular matrix (ECM) and cell-to-cell interactions and enhancing paracrine effect. Furthermore, the sustained release of PDGF-BB can recruit endogenous MSCs to prolong the paracrine activity of the living GMs. In vitro and in vivo experiments validated that the living GMs exhibit superior secretion properties and anti-inflammatory efficacy and can attenuate osteoarthritis (OA) progression by favoring the adherent microenvironment and utilizing the synergistic effect of exogenous and endogenous MSCs. Overall, a living injectable porous hydrogel microsphere that can enhance the paracrine activity of stem cells is fabricated and anticipated to hold the potential of future clinical translation in OA and other diseases.
Collapse
Affiliation(s)
- Xingchen Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiaoxiao Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Jielai Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Jiawei Lin
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yuan Zhu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Xiangyang Xu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
14
|
Lopes-Pacheco M, Rocco PRM. Functional enhancement strategies to potentiate the therapeutic properties of mesenchymal stromal cells for respiratory diseases. Front Pharmacol 2023; 14:1067422. [PMID: 37007034 PMCID: PMC10062457 DOI: 10.3389/fphar.2023.1067422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Respiratory diseases remain a major health concern worldwide because they subject patients to considerable financial and psychosocial burdens and result in a high rate of morbidity and mortality. Although significant progress has been made in understanding the underlying pathologic mechanisms of severe respiratory diseases, most therapies are supportive, aiming to mitigate symptoms and slow down their progressive course but cannot improve lung function or reverse tissue remodeling. Mesenchymal stromal cells (MSCs) are at the forefront of the regenerative medicine field due to their unique biomedical potential in promoting immunomodulation, anti-inflammatory, anti-apoptotic and antimicrobial activities, and tissue repair in various experimental models. However, despite several years of preclinical research on MSCs, therapeutic outcomes have fallen far short in early-stage clinical trials for respiratory diseases. This limited efficacy has been associated with several factors, such as reduced MSC homing, survival, and infusion in the late course of lung disease. Accordingly, genetic engineering and preconditioning methods have emerged as functional enhancement strategies to potentiate the therapeutic actions of MSCs and thus achieve better clinical outcomes. This narrative review describes various strategies that have been investigated in the experimental setting to functionally potentiate the therapeutic properties of MSCs for respiratory diseases. These include changes in culture conditions, exposure of MSCs to inflammatory environments, pharmacological agents or other substances, and genetic manipulation for enhanced and sustained expression of genes of interest. Future directions and challenges in efficiently translating MSC research into clinical practice are discussed.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| |
Collapse
|
15
|
Li J, Li K, Du Y, Tang X, Liu C, Cao S, Zhao B, Huang H, Zhao H, Kong W, Xu T, Shao C, Shao J, Zhang G, Lan H, Xi Y. Dual-Nozzle 3D Printed Nano-Hydroxyapatite Scaffold Loaded with Vancomycin Sustained-Release Microspheres for Enhancing Bone Regeneration. Int J Nanomedicine 2023; 18:307-322. [PMID: 36700146 PMCID: PMC9868285 DOI: 10.2147/ijn.s394366] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/24/2022] [Indexed: 01/19/2023] Open
Abstract
Background Successful treatment of infectious bone defect remains a major challenge in the orthopaedic field. At present, the conventional treatment for infectious bone defects is surgical debridement and long-term systemic antibiotic use. It is necessary to develop a new strategy to achieve effective bone regeneration and local anti-infection for infectious bone defects. Methods Firstly, vancomycin / poly (lactic acid-glycolic acid) sustained release microspheres (VAN/PLGA-MS) were prepared. Then, through the dual-nozzle 3D printing technology, VAN/PLGA-MS was uniformly loaded into the pores of nano-hydroxyapatite (n-HA) and polylactic acid (PLA) scaffolds printed in a certain proportion, and a composite scaffold (VAN/MS-PLA/n-HA) was designed, which can not only promote bone repair but also resist local infection. Finally, the performance of the composite scaffold was evaluated by in vivo and in vitro biological evaluation. Results The in vitro release test of microspheres showed that the release of VAN/PLGA-MS was relatively stable from the second day, and the average daily release concentration was about 15.75 μg/mL, which was higher than the minimum concentration specified in the guidelines. The bacteriostatic test in vitro showed that VAN/PLGA-MS had obvious inhibitory effect on Staphylococcus aureus ATCC-29213. Biological evaluation of VAN/MS-PLA/n-HA scaffolds in vitro showed that it can promote the proliferation of adipose stem cells. In vivo biological evaluation showed that VAN/MS-PLA/n-HA scaffold could significantly promote bone regeneration. Conclusion Our research shows that VAN/MS-PLA/n-HA scaffolds have satisfying biomechanical properties, effectively inhibit the growth of Staphylococcus aureus, with good biocompatibility, and effectiveness on repairing bone defects. The VAN/MS-PLA/n-HA scaffold provide the clinic with an application prospect in bone tissue engineering.
Collapse
Affiliation(s)
- Jianyi Li
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Keke Li
- Yantai Campus of Binzhou Medical University, Yantai, People’s Republic of China
| | - Yukun Du
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Xiaojie Tang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, People’s Republic of China
| | - Chenjing Liu
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Shannan Cao
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, People’s Republic of China
| | - Baomeng Zhao
- Yantai Campus of Binzhou Medical University, Yantai, People’s Republic of China
| | - Hai Huang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, People’s Republic of China
| | - Hongri Zhao
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, People’s Republic of China
| | - Weiqing Kong
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Tongshuai Xu
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, People’s Republic of China
| | - Cheng Shao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Jiale Shao
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Guodong Zhang
- Tengzhou Central People’s Hospital, Tengzhou, People’s Republic of China
| | - Hongbo Lan
- Shandong Engineering Research Center for Additive Manufacturing Qingdao University of Technology, Qingdao, People’s Republic of China,Hongbo Lan, Shandong Engineering Research Center for Additive Manufacturing Qingdao University of Technology, Qingdao, 266520, People’s Republic of China, Email
| | - Yongming Xi
- Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China,Correspondence: Yongming Xi, Department of Orthopaedic Surgery, the Affiliated Hospital of Qingdao University, Qingdao, 266071, People’s Republic of China, Email
| |
Collapse
|
16
|
Chinnadurai R. Advanced Technologies for Potency Assay Measurement. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:81-95. [PMID: 37258785 DOI: 10.1007/978-3-031-30040-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Crucial for their application, cell products need to be well-characterized in the cell manufacturing facilities and conform to regulatory approval criteria before infusion into the patients. Mesenchymal Stromal Cells (MSCs) are the leading cell therapy candidate in clinical trials worldwide. Early phase clinical trials have demonstrated that MSCs display an excellent safety profile and are well tolerated. However, MSCs have also exhibited contradictory efficacy in later-phase clinical trials with reasons for this discrepancy including poorly understood mechanism of MSC therapeutic action. With likelihood that a number of attributes are involved in MSC derived clinical benefit, an assay that measures a single quality of may not adequately reflect potency, thus a combination of bioassays and analytical methods, collectively called "assay matrix" are favoured for defining the potency of MSC more adequately. This chapter highlights advanced technologies and targets that can achieve quantitative measurement for a range of MSC attributes, including immunological, genomic, secretome, phosphorylation, morphological, biomaterial, angiogenic and metabolic assays.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA.
| |
Collapse
|
17
|
Niebergall-Roth E, Frank NY, Ganss C, Frank MH, Kluth MA. Skin-Derived ABCB5 + Mesenchymal Stem Cells for High-Medical-Need Inflammatory Diseases: From Discovery to Entering Clinical Routine. Int J Mol Sci 2022; 24:66. [PMID: 36613507 PMCID: PMC9820160 DOI: 10.3390/ijms24010066] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
The ATP-binding cassette superfamily member ABCB5 identifies a subset of skin-resident mesenchymal stem cells (MSCs) that exhibit potent immunomodulatory and wound healing-promoting capacities along with superior homing ability. The ABCB5+ MSCs can be easily accessed from discarded skin samples, expanded, and delivered as a highly homogenous medicinal product with standardized potency. A range of preclinical studies has suggested therapeutic efficacy of ABCB5+ MSCs in a variety of currently uncurable skin and non-skin inflammatory diseases, which has been substantiated thus far by distinct clinical trials in chronic skin wounds or recessive dystrophic epidermolysis bullosa. Therefore, skin-derived ABCB5+ MSCs have the potential to provide a breakthrough at the forefront of MSC-based therapies striving to fulfill current unmet medical needs. The most recent milestones in this regard are the approval of a phase III pivotal trial of ABCB5+ MSCs for treatment of recessive dystrophic and junctional epidermolysis bullosa by the US Food and Drug Administration, and national market access of ABCB5+ MSCs (AMESANAR®) for therapy-refractory chronic venous ulcers under the national hospital exemption pathway in Germany.
Collapse
Affiliation(s)
| | - Natasha Y. Frank
- Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christoph Ganss
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| | - Markus H. Frank
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
- Transplant Research Program, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- School of Medical and Health Sciences, Edith Cowan University, Perth 6027, Australia
| | - Mark A. Kluth
- TICEBA GmbH, 69120 Heidelberg, Germany
- RHEACELL GmbH & Co. KG, 69120 Heidelberg, Germany
| |
Collapse
|
18
|
Silva-Carvalho AÉ, da Silva IGM, Corrêa JR, Saldanha-Araujo F. Regulatory T-Cell Enhancement, Expression of Adhesion Molecules, and Production of Anti-Inflammatory Factors Are Differentially Modulated by Spheroid-Cultured Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:ijms232214349. [PMID: 36430835 PMCID: PMC9695986 DOI: 10.3390/ijms232214349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
The culture of mesenchymal stem cells (MSCs) as spheroids promotes a more physiological cellular behavior, as it more accurately reflects the biological microenvironment. Nevertheless, mixed results have been found regarding the immunosuppressive properties of spheroid-cultured MSCs (3D-MSCs), the mechanisms of immunoregulation of 3D-MSCs being scarcely described at this point. In the present study, we constructed spheroids from MSCs and compared their immunosuppressive potential with that of MSCs cultured in monolayer (2D-MSCs). First, we evaluated the ability of 2D-MSCs and 3D-MSCs to control the activation and proliferation of T-cells. Next, we evaluated the percentage of regulatory T-cells (Tregs) after the co-culturing of peripheral blood mononuclear cells (PBMCs) with 2D-MSCs and 3D-MSCs. Finally, we investigated the expression of adhesion molecules, as well as the expressions of several anti-inflammatory transcripts in 2D-MSCs and 3D-MSCs maintained in both inflammatory and non-inflammatory conditions. Interestingly, our data show that several anti-inflammatory genes are up-regulated in 3D-MSCs, and that these cells can control T-cell proliferation. Nevertheless, 2D-MSCs are more efficient in suppressing the immune cell proliferation. Importantly, contrary to what was observed in 3D-MSCs, the expressions of ICAM-1 and VCAM-1 are significantly upregulated in 2D-MSCs exposed to an inflammatory environment. Furthermore, only 2D-MSCs are able to promote the enhancement of Tregs. Taken together, our data clearly show that the immunosuppressive potential of MSCs is significantly impacted by their shape, and highlights the important role of cell-cell adhesion molecules for optimal MSC immunomodulatory function.
Collapse
Affiliation(s)
- Amandda Évelin Silva-Carvalho
- Hematology and Stem Cells Laboratory, University of Brasília, Brasilia 70910-900, Brazil
- Molecular Pharmacology Laboratory, University of Brasília, Brasilia 70910-900, Brazil
| | | | - José Raimundo Corrêa
- Microscopy and Microanalysis Laboratory, University of Brasília, Brasilia 70910-900, Brazil
| | - Felipe Saldanha-Araujo
- Hematology and Stem Cells Laboratory, University of Brasília, Brasilia 70910-900, Brazil
- Correspondence: ; Tel./Fax: +55-61-3107-2008
| |
Collapse
|
19
|
Shephard MT, Merkhan MM, Forsyth NR. Human Mesenchymal Stem Cell Secretome Driven T Cell Immunomodulation Is IL-10 Dependent. Int J Mol Sci 2022; 23:13596. [PMID: 36362383 PMCID: PMC9658100 DOI: 10.3390/ijms232113596] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 09/01/2023] Open
Abstract
The Human Mesenchymal Stem Cell (hMSC) secretome has pleiotropic effects underpinning its therapeutic potential. hMSC serum-free conditioned media (SFCM) contains a variety of cytokines, with previous studies linking a changed secretome composition to physoxia. The Jurkat T cell model allowed the efficacy of SFCM vs. serum-free media (SFM) in the suppression of immunological aspects, including proliferation and polarisation, to be explored. Cell growth in SFM was higher [(21% O2 = 5.3 × 105 ± 1.8 × 104 cells/mL) and (2% O2 = 5.1 × 105 ± 3.0 × 104 cells/mL)], compared to SFCM [(21% O2 = 2.4 × 105 ± 2.5 × 104 cells/mL) and (2% O2 = 2.2 × 105 ± 5.8 × 103 cells/mL)]. SFM supported IL-2 release following activation [(21% O2 = 5305 ± 211 pg/mL) and (2% O2 = 5347 ± 327 pg/mL)] whereas SFCM suppressed IL-2 secretion [(21% O2 = 2461 ± 178 pg/mL) and (2% O2 = 1625 ± 159 pg/mL)]. Anti-inflammatory cytokines, namely IL-4, IL-10, and IL-13, which we previously confirmed as components of hMSC SFCM, were tested. IL-10 neutralisation in SFCM restored proliferation in both oxygen environments (SFM/SFCM+antiIL-10 ~1-fold increase). Conversely, IL-4/IL-13 neutralisation showed no proliferation restoration [(SFM/SFM+antiIL-4 ~2-fold decrease), and (SFM/SFCM+antiIL-13 ~2-fold decrease)]. Present findings indicate IL-10 played an immunosuppressive role by reducing IL-2 secretion. Identification of immunosuppressive components of the hMSC secretome and a mechanistic understanding of their action allow for the advancement and refinement of potential future cell-free therapies.
Collapse
Affiliation(s)
- Matthew T. Shephard
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, UK
| | - Marwan M. Merkhan
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, UK
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Mosul, Mosul 41002, Iraq
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Staffordshire ST4 7QB, UK
| |
Collapse
|
20
|
Allen-Coyle TJ, Niu J, Welsch E, Conlon NT, Garner W, Clynes M, O'Sullivan F, Straubinger RM, Mager DE, Roche S. FOLFIRINOX Pharmacodynamic Interactions in 2D and 3D Pancreatic Cancer Cell Cultures. AAPS J 2022; 24:108. [PMID: 36229752 DOI: 10.1208/s12248-022-00752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
The multi-drug combination regime, FOLFIRINOX, is a standard of care chemotherapeutic therapy for pancreatic cancer patients. However, systematic evaluation of potential pharmacodynamic interactions among multi-drug therapy has not been reported previously. Here, pharmacodynamic interactions of the FOLFIRINOX agents (5-fluorouracil (5-FU), oxaliplatin (Oxa) and SN-38, the active metabolite of irinotecan) were assessed across a panel of primary and established pancreatic cancer cells. Inhibition of cell proliferation was quantified for each drug, alone and in combination, to obtain quantitative, drug-specific interaction parameters and assess the nature of drug interactions. The experimental data were analysed assuming Bliss independent interactions, and nonlinear regression model fitting was conducted in SAS. Estimates of the drug interaction term, psi (ψ), revealed that the Oxa/SN-38 combination appeared synergistic in PANC-1 (ψ = 0.6, 95% CI = 0.4, 0.9) and modestly synergistic, close to additive, in MIAPaCa-2 (ψ = 0.8, 95% CI = 0.6, 1.0) in 2D assays. The triple combination was strongly synergistic in MIAPaCa-2 (ψ = 0.2, 95% CI = 0.1, 0.3) and modestly synergistic/borderline additive in PANC-1 2D (ψ = 0.8, 95% CI = 0.6, 1.0). The triple combination showed antagonistic interactions in the primary PIN-127 and 3D PANC-1 model (ψ > 1). Quantitative pharmacodynamic interactions have not been described for the FOLFIRINOX regimen; this analysis suggests a complex interplay among the three chemotherapeutic agents. Extension of this pharmacodynamic analysis approach to clinical/translational studies of the FOLFIRINOX combination could reveal additional pharmacodynamic interactions and guide further refinement of this regimen to achieve optimal clinical responses.
Collapse
Affiliation(s)
- Taylor J Allen-Coyle
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland. .,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.
| | - Jin Niu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, New York, Albany, USA
| | - Eva Welsch
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland
| | - Neil T Conlon
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland
| | - Weylon Garner
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, New York, Albany, USA
| | - Martin Clynes
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland.,Pancreatic Cancer Research Fund UK (PCRF), London, UK
| | - Finbarr O'Sullivan
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland.,National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, New York, Albany, USA.,Departments of Pharmacology & Therapeutics, and Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, New York, Albany, USA.,Enhanced Pharmacodynamics, LLC, Buffalo, New York, USA
| | - Sandra Roche
- SSPC, The SFI Research Centre for Pharmaceuticals, Limerick, Ireland
| |
Collapse
|
21
|
Shi L, Zhang Z, Deng M, Zheng F, Liu W, Ye S. Biological mechanisms and applied prospects of mesenchymal stem cells in premature ovarian failure. Medicine (Baltimore) 2022; 101:e30013. [PMID: 35960112 PMCID: PMC9371578 DOI: 10.1097/md.0000000000030013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/05/2022] [Accepted: 06/06/2022] [Indexed: 01/04/2023] Open
Abstract
Premature ovarian failure (POF), also known as primary ovarian insufficiency (POI), refers to the loss of ovarian function in women after puberty and before the age of 40 characterized by high serum gonadotropins and low estrogen, irregular menstruation, amenorrhea, and decreased fertility. However, the specific pathogenesis of POF is unexplained, and there is no effective therapy for its damaged ovarian tissue structure and reduced reserve function. Mesenchymal stem cells (MSCs), with multidirectional differentiation potential and self-renewal ability, as well as the cytokines and exosomes they secrete, have been studied and tested to play an active therapeutic role in a variety of degenerative pathologies, and MSCs are the most widely used stem cells in regenerative medicine. MSCs can reverse POI and enhance ovarian reserve function through differentiation into granulosa cells (GCs), immune regulation, secretion of cytokines and other nutritional factors, reduction of GCs apoptosis, and promotion of GCs regeneration. Many studies have proved that MSCs may have a restorative effect on the structure and fertility of injured ovarian tissues and turn to be a useful clinical approach to the treatment of patients with POF in recent years. We intend to use MSCs-based therapy to completely reverse POI in the future.
Collapse
Affiliation(s)
- Lan Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zhifen Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Miao Deng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Fangyuan Zheng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Wenhua Liu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Shujin Ye
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
22
|
Islam S, Parker J, Dash BC, Hsia HC. Human iPSC-Vascular smooth muscle cell spheroids demonstrate size-dependent alterations in cellular viability and secretory function. J Biomed Mater Res A 2022; 110:1813-1823. [PMID: 35815599 DOI: 10.1002/jbm.a.37423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 11/11/2022]
Abstract
Human-induced pluripotent stem cells (hiPSC) and their differentiated vascular cells have been revolutionizing the field of regenerative wound healing. These cells are shown to be rejuvenated with immense potentials in secreting paracrine factors. Recently, hiPSC-derived vascular smooth muscle cells (hiPSC-VSMC) have shown regenerative wound healing ability via their paracrine secretion. The quest to modulate the secretory function of these hiPSC-VSMC is an ongoing effort and involves the use of both biochemical and biophysical stimuli. This study explores the development and optimization of a reproducible, inexpensive protocol to form hiPSC-VSMC derived spheroids to investigate the implications of spheroid size on viability and paracrine secretion. Our data show the successful formation of different sizes of spheroids using various amount of hiPSC-VSMC. The hiPSC-VSMC spheroids formed with 10,000 cells strike an ideal balance between overall cell health and maximal paracrine secretion. The conditioned medium from these spheroids was found to be bioactive in enhancing human dermal fibroblast cell proliferation and migration. This research will inform future studies on the optimal spheroid size for regenerative wound healing applications.
Collapse
Affiliation(s)
- Sara Islam
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Jackson Parker
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Biraja C Dash
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Henry C Hsia
- Section of Plastic Surgery, Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, USA.,Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
23
|
Vackova I, Vavrinova E, Musilkova J, Havlas V, Petrenko Y. Hypothermic Storage of 3D Cultured Multipotent Mesenchymal Stromal Cells for Regenerative Medicine Applications. Polymers (Basel) 2022; 14:polym14132553. [PMID: 35808601 PMCID: PMC9269598 DOI: 10.3390/polym14132553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 02/01/2023] Open
Abstract
The regulatory requirements in cell processing, in the choice of a biomaterial scaffold and in quality control analysis, have to be followed in the clinical application of tissue-engineered grafts. Confirmation of sterility during quality control studies requires prolonged storage of the cell-based construct. After storage, preservation of the functional properties of the cells is an important prerequisite if the cells are to be used for cell-based tissue therapies. The study presented here shows the generation of 3D constructs based on Wharton’s jelly multipotent mesenchymal stromal cells (WJ-MSCs) and the clinically-acceptable HyaloFast® scaffold, and the effect of two- and six-day hypothermic storage of 3D cell-based constructs on the functional properties of populated cells. To study the viability, growth, gene expression, and paracrine secretion of WJ-MSCs within the scaffolds before and after storage, xeno-free culture conditions, metabolic, qPCR, and multiplex assays were applied. The WJ-MSCs adhered and proliferated within the 3D HyaloFast®. Our results show different viability of the cells after the 3D constructs have been stored under mild (25 °C) or strong (4 °C) hypothermia. At 4 °C, the significant decrease of metabolic activity of WJ-MSCs was detected after 2 days of storage, with almost complete cell loss after 6 days. In mild hypothermia (25 °C) the decrease in metabolic activity was less remarkable, confirming the suitability of these conditions for cell preservation in 3D environment. The significant changes were detected in gene expression and in the paracrine secretion profile after 2 and 6 days of storage at 25 °C. The results presented in this study are important for the rapid transfer of tissue engineering approaches into clinical applications.
Collapse
Affiliation(s)
- Irena Vackova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of Czech Academy of Science, 14220 Prague, Czech Republic; (I.V.); (J.M.)
| | - Eliska Vavrinova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Jana Musilkova
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of Czech Academy of Science, 14220 Prague, Czech Republic; (I.V.); (J.M.)
| | - Vojtech Havlas
- Department of Orthopaedics and Traumatology, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic;
| | - Yuriy Petrenko
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of Czech Academy of Science, 14220 Prague, Czech Republic; (I.V.); (J.M.)
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
- Correspondence:
| |
Collapse
|
24
|
Olmedo-Moreno L, Aguilera Y, Baliña-Sánchez C, Martín-Montalvo A, Capilla-González V. Heterogeneity of In Vitro Expanded Mesenchymal Stromal Cells and Strategies to Improve Their Therapeutic Actions. Pharmaceutics 2022; 14:1112. [PMID: 35631698 PMCID: PMC9146397 DOI: 10.3390/pharmaceutics14051112] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 12/12/2022] Open
Abstract
Beneficial properties of mesenchymal stromal cells (MSCs) have prompted their use in preclinical and clinical research. Accumulating evidence has been provided for the therapeutic effects of MSCs in several pathologies, including neurodegenerative diseases, myocardial infarction, skin problems, liver disorders and cancer, among others. Although MSCs are found in multiple tissues, the number of MSCs is low, making in vitro expansion a required step before MSC application. However, culture-expanded MSCs exhibit notable differences in terms of cell morphology, physiology and function, which decisively contribute to MSC heterogeneity. The changes induced in MSCs during in vitro expansion may account for the variability in the results obtained in different MSC-based therapy studies, including those using MSCs as living drug delivery systems. This review dissects the different changes that occur in culture-expanded MSCs and how these modifications alter their therapeutic properties after transplantation. Furthermore, we discuss the current strategies developed to improve the beneficial effects of MSCs for successful clinical implementation, as well as potential therapeutic alternatives.
Collapse
Affiliation(s)
| | | | | | | | - Vivian Capilla-González
- Department of Regeneration and Cell Therapy, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER)-CSIC-US-UPO, 41092 Seville, Spain; (L.O.-M.); (Y.A.); (C.B.-S.); (A.M.-M.)
| |
Collapse
|
25
|
Li M, Jiang Y, Hou Q, Zhao Y, Zhong L, Fu X. Potential pre-activation strategies for improving therapeutic efficacy of mesenchymal stem cells: current status and future prospects. Stem Cell Res Ther 2022; 13:146. [PMID: 35379361 PMCID: PMC8981790 DOI: 10.1186/s13287-022-02822-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/20/2022] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cell (MSC)-based therapy has been considered as a promising approach targeting a variety of intractable diseases due to remarkable multiple effect of MSCs, such as multilineage differentiation, immunomodulatory property, and pro-regenerative capacity. However, poor engraftment, low survival rate of transplanted MSC, and impaired donor-MSC potency under host age/disease result in unsatisfactory therapeutic outcomes. Enhancement strategies, including genetic manipulation, pre-activation, and modification of culture method, have been investigated to generate highly functional MSC, and approaches for MSC pre-activation are highlighted. In this review, we summarized the current approaches of MSC pre-activation and further classified, analysed the scientific principles and main characteristics of these manipulations, and described the pros and cons of individual pre-activation strategies. We also discuss the specialized tactics to solve the challenges in this promising field so that it improves MSC therapeutic functions to serve patients better.
Collapse
Affiliation(s)
- Meirong Li
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| | - Yufeng Jiang
- Wound Repairing Department, PLA Strategic Support Force Characteristic Medical Center, Beijing, 100101, China
| | - Qian Hou
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Yali Zhao
- Central Laboratory, Trauma Treatment Center, Chinese PLA General Hospital, Hainan Hospital, Sanya, China
| | - Lingzhi Zhong
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China.,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and 4th Medical Center, PLA General Hospital and PLA Medical College, Beijing, China. .,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration, Beijing, China. .,Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences 2019RU051, Beijing, China.
| |
Collapse
|
26
|
Khetan S. Cryopreservation of network bioactivity and multi-lineage stromal cell differentiation potential within three-dimensional synthetic hydrogels. Cryobiology 2021; 105:41-49. [PMID: 34922883 DOI: 10.1016/j.cryobiol.2021.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/30/2022]
Abstract
Despite progress in many aspects of controlling cell behavior within synthetic three-dimensional hydrogels, approaches to cryopreserve these systems - encompassing the protection of both encapsulated cell viability and network bioactive functions - are lacking. Here, we demonstrate the retention of encapsulated human mesenchymal stromal cell (hMSC) viability following in situ cryopreservation regardless of cell line, material system, or storage duration. Further, the preservation extends to network bioactive functions, with hMSCs cryopreserved within degradable and adhesive hyaluronic-acid (HA) based hydrogels exhibiting degradation-mediated spreading within the gels equivalent to their non-frozen counterparts. Finally, the platform cryopreservation protocol preserves multi-lineage cellular differentiation capacity, with encapsulated hMSCs in non-degradable and adhesive/degradable HA-based hydrogels undergoing rates of adipogenesis and osteogenesis, respectively, equivalent to those in non-frozen gels on a per-cell basis. Collectively, these findings indicate a versatile platform technology that contributes to an increased understanding of three-dimensional cell-matrix interactions, and which may enable the indefinite cryopreservation of tissue engineering constructs for clinical applications.
Collapse
Affiliation(s)
- Sudhir Khetan
- Department of Electrical, Computer, and Biomedical Engineering, Union College, Schenectady, NY, USA.
| |
Collapse
|
27
|
Pereira DR, Silva-Correia J, Oliveira JM, Reis RL, Pandit A. Macromolecular modulation of a 3D hydrogel construct differentially regulates human stem cell tissue-to-tissue interface. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 133:112611. [DOI: 10.1016/j.msec.2021.112611] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/25/2021] [Accepted: 12/11/2021] [Indexed: 01/21/2023]
|
28
|
Shanbhag S, Kampleitner C, Mohamed-Ahmed S, Yassin MA, Dongre H, Costea DE, Tangl S, Hassan MN, Stavropoulos A, Bolstad AI, Suliman S, Mustafa K. Ectopic Bone Tissue Engineering in Mice Using Human Gingiva or Bone Marrow-Derived Stromal/Progenitor Cells in Scaffold-Hydrogel Constructs. Front Bioeng Biotechnol 2021; 9:783468. [PMID: 34917602 PMCID: PMC8670384 DOI: 10.3389/fbioe.2021.783468] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/16/2021] [Indexed: 01/22/2023] Open
Abstract
Three-dimensional (3D) spheroid culture can promote the osteogenic differentiation and bone regeneration capacity of mesenchymal stromal cells (MSC). Gingiva-derived progenitor cells (GPC) represent a less invasive alternative to bone marrow MSC (BMSC) for clinical applications. The aim of this study was to test the in vivo bone forming potential of human GPC and BMSC cultured as 3D spheroids or dissociated cells (2D). 2D and 3D cells encapsulated in constructs of human platelet lysate hydrogels (HPLG) and 3D-printed poly (L-lactide-co-trimethylene carbonate) scaffolds (HPLG-PLATMC) were implanted subcutaneously in nude mice; cell-free HPLG-PLATMC constructs served as a control. Mineralization was assessed using micro-computed tomography (µCT), histology, scanning electron microscopy (SEM) and in situ hybridization (ISH). After 4–8 weeks, µCT revealed greater mineralization in 3D-BMSC vs. 2D-BMSC and 3D-GPC (p < 0.05), and a similar trend in 2D-GPC vs. 2D-BMSC (p > 0.05). After 8 weeks, greater mineralization was observed in cell-free constructs vs. all 2D- and 3D-cell groups (p < 0.05). Histology and SEM revealed an irregular but similar mineralization pattern in all groups. ISH revealed similar numbers of 2D and 3D BMSC/GPC within and/or surrounding the mineralized areas. In summary, spheroid culture promoted ectopic mineralization in constructs of BMSC, while constructs of dissociated GPC and BMSC performed similarly. The combination of HPLG and PLATMC represents a promising scaffold for bone tissue engineering applications.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
- *Correspondence: Siddharth Shanbhag, ; Kamal Mustafa,
| | - Carina Kampleitner
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVA, Vienna, Austria
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Samih Mohamed-Ahmed
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Mohammed Ahmad Yassin
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Harsh Dongre
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Daniela Elena Costea
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Stefan Tangl
- Karl Donath Laboratory for Hard Tissue and Biomaterial Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mohamad Nageeb Hassan
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Andreas Stavropoulos
- Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Anne Isine Bolstad
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Salwa Suliman
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
- *Correspondence: Siddharth Shanbhag, ; Kamal Mustafa,
| |
Collapse
|
29
|
Shanbhag S, Suliman S, Mohamed-Ahmed S, Kampleitner C, Hassan MN, Heimel P, Dobsak T, Tangl S, Bolstad AI, Mustafa K. Bone regeneration in rat calvarial defects using dissociated or spheroid mesenchymal stromal cells in scaffold-hydrogel constructs. Stem Cell Res Ther 2021; 12:575. [PMID: 34776000 PMCID: PMC8591809 DOI: 10.1186/s13287-021-02642-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/22/2021] [Indexed: 12/20/2022] Open
Abstract
Background Three-dimensional (3D) spheroid culture can promote the osteogenic differentiation of bone marrow mesenchymal stromal cells (BMSC). 3D printing offers the possibility to produce customized scaffolds for complex bone defects. The aim of this study was to compare the potential of human BMSC cultured as 2D monolayers or 3D spheroids encapsulated in constructs of 3D-printed poly-L-lactide-co-trimethylene carbonate scaffolds and modified human platelet lysate hydrogels (PLATMC-HPLG) for bone regeneration. Methods PLATMC-HPLG constructs with 2D or 3D BMSC were assessed for osteogenic differentiation based on gene expression and in vitro mineralization. Subsequently, PLATMC-HPLG constructs with 2D or 3D BMSC were implanted in rat calvarial defects for 12 weeks; cell-free constructs served as controls. Bone regeneration was assessed via in vivo computed tomography (CT), ex vivo micro-CT and histology. Results Osteogenic gene expression was significantly enhanced in 3D versus 2D BMSC prior to, but not after, encapsulation in PLATMC-HPLG constructs. A trend for greater in vitro mineralization was observed in constructs with 3D versus 2D BMSC (p > 0.05). In vivo CT revealed comparable bone formation after 4, 8 and 12 weeks in all groups. After 12 weeks, micro-CT revealed substantial regeneration in 2D BMSC (62.47 ± 19.46%), 3D BMSC (51.01 ± 24.43%) and cell-free PLATMC-HPLG constructs (43.20 ± 30.09%) (p > 0.05). A similar trend was observed in the histological analysis. Conclusion Despite a trend for superior in vitro mineralization, constructs with 3D and 2D BMSC performed similarly in vivo. Regardless of monolayer or spheroid cell culture, PLATMC-HPLG constructs represent promising scaffolds for bone tissue engineering applications. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02642-w.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway. .,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway.
| | - Salwa Suliman
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Samih Mohamed-Ahmed
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Carina Kampleitner
- Core Facility Hard Tissue and Biomaterial Research/Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Traumatology, The research center in cooperation with AUVA, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mohamed Nageeb Hassan
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Patrick Heimel
- Core Facility Hard Tissue and Biomaterial Research/Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Traumatology, The research center in cooperation with AUVA, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Toni Dobsak
- Core Facility Hard Tissue and Biomaterial Research/Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Stefan Tangl
- Core Facility Hard Tissue and Biomaterial Research/Karl Donath Laboratory, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Anne Isine Bolstad
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway
| | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, 5009, Bergen, Norway.
| |
Collapse
|
30
|
Modulation of Inherent Niches in 3D Multicellular MSC Spheroids Reconfigures Metabolism and Enhances Therapeutic Potential. Cells 2021; 10:cells10102747. [PMID: 34685727 PMCID: PMC8534378 DOI: 10.3390/cells10102747] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 01/02/2023] Open
Abstract
Multicellular spheroids show three-dimensional (3D) organization with extensive cell–cell and cell–extracellular matrix interactions. Owing to their native tissue-mimicking characteristics, mesenchymal stem cell (MSC) spheroids are considered promising as implantable therapeutics for stem cell therapy. Herein, we aim to further enhance their therapeutic potential by tuning the cultivation parameters and thus the inherent niche of 3D MSC spheroids. Significantly increased expression of multiple pro-regenerative paracrine signaling molecules and immunomodulatory factors by MSCs was observed after optimizing the conditions for spheroid culture. Moreover, these alterations in cellular behaviors may be associated with not only the hypoxic niche developed in the spheroid core but also with the metabolic reconfiguration of MSCs. The present study provides efficient methods for manipulating the therapeutic capacity of 3D MSC spheroids, thus laying solid foundations for future development and clinical application of spheroid-based MSC therapy for regenerative medicine.
Collapse
|
31
|
Shanbhag S, Rashad A, Nymark EH, Suliman S, de Lange Davies C, Stavropoulos A, Bolstad AI, Mustafa K. Spheroid Coculture of Human Gingiva-Derived Progenitor Cells With Endothelial Cells in Modified Platelet Lysate Hydrogels. Front Bioeng Biotechnol 2021; 9:739225. [PMID: 34513817 PMCID: PMC8427051 DOI: 10.3389/fbioe.2021.739225] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/12/2021] [Indexed: 01/12/2023] Open
Abstract
Cell coculture strategies can promote angiogenesis within tissue engineering constructs. This study aimed to test the angiogenic potential of human umbilical vein endothelial cells (HUVEC) cocultured with gingiva-derived progenitor cells (GPC) as spheroids in a xeno-free environment. Human platelet lysate (HPL) was used as a cell culture supplement and as a hydrogel matrix (HPLG) for spheroid encapsulation. HUVEC and HUVEC + GPC (1:1 or 5:1) spheroids were encapsulated in various HPLG formulations. Angiogenesis was assessed via in vitro sprouting and in vivo chick chorioallantoic membrane (CAM) assays. HUVEC revealed characteristic in vitro sprouting in HPL/HPLG and this was significantly enhanced in cocultures with GPC (p < 0.05). A trend for greater sprouting was observed in 5:1 vs 1:1 HUVEC + GPC spheroids and in certain HPLG formulations (p > 0.05). Both HUVEC and HUVEC + GPC spheroids in HPLG revealed abundant and comparable neoangiogenesis in the CAM assay (p > 0.05). Spheroid coculture of HUVEC + GPC in HPLG represents a promising strategy to promote angiogenesis.
Collapse
Affiliation(s)
- Siddharth Shanbhag
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.,Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Ahmad Rashad
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Ellen Helgeland Nymark
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Salwa Suliman
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | | | - Andreas Stavropoulos
- Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden.,Division of Regenerative Medicine and Periodontology, University Clinics of Dental Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Isine Bolstad
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Center for Translational Oral Research (TOR), Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
32
|
Bioprintable Lung Extracellular Matrix Hydrogel Scaffolds for 3D Culture of Mesenchymal Stromal Cells. Polymers (Basel) 2021; 13:polym13142350. [PMID: 34301107 PMCID: PMC8309540 DOI: 10.3390/polym13142350] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal cell (MSC)-based cell therapy in acute respiratory diseases is based on MSC secretion of paracrine factors. Several strategies have proposed to improve this are being explored including pre-conditioning the MSCs prior to administration. We here propose a strategy for improving the therapeutic efficacy of MSCs based on cell preconditioning by growing them in native extracellular matrix (ECM) derived from the lung. To this end, a bioink with tunable stiffness based on decellularized porcine lung ECM hydrogels was developed and characterized. The bioink was suitable for 3D culturing of lung-resident MSCs without the need for additional chemical or physical crosslinking. MSCs showed good viability, and contraction assays showed the existence of cell–matrix interactions in the bioprinted scaffolds. Adhesion capacity and length of the focal adhesions formed were increased for the cells cultured within the lung hydrogel scaffolds. Also, there was more than a 20-fold increase of the expression of the CXCR4 receptor in the 3D-cultured cells compared to the cells cultured in plastic. Secretion of cytokines when cultured in an in vitro model of lung injury showed a decreased secretion of pro-inflammatory mediators for the cells cultured in the 3D scaffolds. Moreover, the morphology of the harvested cells was markedly different with respect to conventionally (2D) cultured MSCs. In conclusion, the developed bioink can be used to bioprint structures aimed to improve preconditioning MSCs for therapeutic purposes.
Collapse
|
33
|
Wang Z, Zhu X, Cong X. Spatial micro-variation of 3D hydrogel stiffness regulates the biomechanical properties of hMSCs. Biofabrication 2021; 13. [PMID: 34107453 DOI: 10.1088/1758-5090/ac0982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are one of the most promising candidates for cell-based therapeutic products. Nonetheless, their biomechanical phenotype afterin vitroexpansion is still unsatisfactory, for example, restricting the efficiency of microcirculation of delivered hMSCs for further cell therapies. Here, we propose a scheme using maleimide-dextran hydrogel with locally varied stiffness in microscale to modify the biomechanical properties of hMSCs in three-dimensional (3D) niches. We show that spatial micro-variation of stiffness can be controllably generated in the hydrogel with heterogeneously cross-linking via atomic force microscopy measurements. The result of 3D cell culture experiment demonstrates the hydrogels trigger the formation of multicellular spheroids, and the derived hMSCs could be rationally softened via adjustment of the stiffness variation (SV) degree. Importantly,in vitro, the hMSCs modified with the higher SV degree can pass easier through capillary-shaped micro-channels. Further, we discuss the underlying mechanics of the increased cellular elasticity by focusing on the effect of rearranged actin networks, via the proposed microscopic model of biomechanically modified cells. Overall, this work highlights the effectiveness of SV-hydrogels in reprogramming and manufacturing hMSCs with designed biomechanical properties for improved therapeutic potential.
Collapse
Affiliation(s)
- Zheng Wang
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiaolu Zhu
- College of Mechanical and Electrical Engineering, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Changzhou Key Laboratory of Digital Manufacture Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China.,Jiangsu Key Laboratory of Special Robot Technology, Hohai University, Changzhou, Jiangsu 213022, People's Republic of China
| | - Xiuli Cong
- Department of Orthopaedics, Zhejiang Hospital, No. 12 Lingyin Road, Hangzhou, Zhejiang 310013, People's Republic of China
| |
Collapse
|
34
|
Sevari SP, Ansari S, Moshaverinia A. A narrative overview of utilizing biomaterials to recapitulate the salient regenerative features of dental-derived mesenchymal stem cells. Int J Oral Sci 2021; 13:22. [PMID: 34193832 PMCID: PMC8245503 DOI: 10.1038/s41368-021-00126-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/26/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering approaches have emerged recently to circumvent many limitations associated with current clinical practices. This elegant approach utilizes a natural/synthetic biomaterial with optimized physiomechanical properties to serve as a vehicle for delivery of exogenous stem cells and bioactive factors or induce local recruitment of endogenous cells for in situ tissue regeneration. Inspired by the natural microenvironment, biomaterials could act as a biomimetic three-dimensional (3D) structure to help the cells establish their natural interactions. Such a strategy should not only employ a biocompatible biomaterial to induce new tissue formation but also benefit from an easily accessible and abundant source of stem cells with potent tissue regenerative potential. The human teeth and oral cavity harbor various populations of mesenchymal stem cells (MSCs) with self-renewing and multilineage differentiation capabilities. In the current review article, we seek to highlight recent progress and future opportunities in dental MSC-mediated therapeutic strategies for tissue regeneration using two possible approaches, cell transplantation and cell homing. Altogether, this paper develops a general picture of current innovative strategies to employ dental-derived MSCs combined with biomaterials and bioactive factors for regenerating the lost or defective tissues and offers information regarding the available scientific data and possible applications.
Collapse
Affiliation(s)
- Sevda Pouraghaei Sevari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sahar Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alireza Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
35
|
Meeremans M, Van de Walle GR, Van Vlierberghe S, De Schauwer C. The Lack of a Representative Tendinopathy Model Hampers Fundamental Mesenchymal Stem Cell Research. Front Cell Dev Biol 2021; 9:651164. [PMID: 34012963 PMCID: PMC8126669 DOI: 10.3389/fcell.2021.651164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Overuse tendon injuries are a major cause of musculoskeletal morbidity in both human and equine athletes, due to the cumulative degenerative damage. These injuries present significant challenges as the healing process often results in the formation of inferior scar tissue. The poor success with conventional therapy supports the need to search for novel treatments to restore functionality and regenerate tissue as close to native tendon as possible. Mesenchymal stem cell (MSC)-based strategies represent promising therapeutic tools for tendon repair in both human and veterinary medicine. The translation of tissue engineering strategies from basic research findings, however, into clinical use has been hampered by the limited understanding of the multifaceted MSC mechanisms of action. In vitro models serve as important biological tools to study cell behavior, bypassing the confounding factors associated with in vivo experiments. Controllable and reproducible in vitro conditions should be provided to study the MSC healing mechanisms in tendon injuries. Unfortunately, no physiologically representative tendinopathy models exist to date. A major shortcoming of most currently available in vitro tendon models is the lack of extracellular tendon matrix and vascular supply. These models often make use of synthetic biomaterials, which do not reflect the natural tendon composition. Alternatively, decellularized tendon has been applied, but it is challenging to obtain reproducible results due to its variable composition, less efficient cell seeding approaches and lack of cell encapsulation and vascularization. The current review will overview pros and cons associated with the use of different biomaterials and technologies enabling scaffold production. In addition, the characteristics of the ideal, state-of-the-art tendinopathy model will be discussed. Briefly, a representative in vitro tendinopathy model should be vascularized and mimic the hierarchical structure of the tendon matrix with elongated cells being organized in a parallel fashion and subjected to uniaxial stretching. Incorporation of mechanical stimulation, preferably uniaxial stretching may be a key element in order to obtain appropriate matrix alignment and create a pathophysiological model. Together, a thorough discussion on the current status and future directions for tendon models will enhance fundamental MSC research, accelerating translation of MSC therapies for tendon injuries from bench to bedside.
Collapse
Affiliation(s)
- Marguerite Meeremans
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Catharina De Schauwer
- Comparative Physiology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
36
|
3D cell sheet structure augments mesenchymal stem cell cytokine production. Sci Rep 2021; 11:8170. [PMID: 33854167 PMCID: PMC8046983 DOI: 10.1038/s41598-021-87571-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/23/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) secrete paracrine factors that play crucial roles during tissue regeneration. An increasing body of evidence suggests that this paracrine function is enhanced by MSC cultivation in three-dimensional (3D) tissue-like microenvironments. Toward this end, this study explored scaffold-free cell sheet technology as a new 3D platform. MSCs cultivated on temperature-responsive culture dishes to a confluent 2D monolayer were harvested by temperature reduction from 37 to 20 °C that induces a surface wettability transition from hydrophobic to hydrophilic. Release of culture-adherent tension induced spontaneous cell sheet contraction, reducing the diameter 2.4-fold, and increasing the thickness 8.0-fold to render a 3D tissue-like construct with a 36% increase in tissue volume. This 2D-to-3D transition reorganized MSC actin cytoskeleton from aligned to multidirectional, corresponding to a cell morphological change from elongated in 2D monolayers to rounded in 3D cell sheets. 3D culture increased MSC gene expression of cell interaction proteins, β-catenin, integrin β1, and connexin 43, and of pro-tissue regenerative cytokines, vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), and interleukin-10 (IL-10), and increased VEGF secretion per MSC 2.1-fold relative to 2D cultures. Together, these findings demonstrate that MSC therapeutic potency can be enhanced by 3D cell sheet tissue structure.
Collapse
|
37
|
Sahu N, Agarwal P, Grand F, Bruschi M, Goodman S, Ammanatullah D, Bhutani N. Encapsulated Mesenchymal Stromal Cell Microbeads Promote Endogenous Regeneration of Osteoarthritic Cartilage Ex Vivo. Adv Healthc Mater 2021; 10:e2002118. [PMID: 33434393 PMCID: PMC10591520 DOI: 10.1002/adhm.202002118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Indexed: 01/01/2023]
Abstract
The anti-inflammatory secretome of mesenchymal stromal cells (MSCs) is lucrative for the treatment of osteoarthritis (OA), a disease characterized by low-grade inflammation. However, the precise effects of the MSC secretome on patient-derived OA tissue is lacking. To investigate these effects, alginate encapsulated MSCs are co-cultured with patient-derived OA cartilage explants for 8 days. Proteoglycan distribution in OA cartilage explants examined by Safranin O staining is markedly improved when cultured with MSC microbeads as compared to control OA explants cultured alone. Total sulfated glycosaminoglycan (sGAG) content in OA explants is significantly increased upon co-culture with MSC microbeads on day 8. The sGAG released into the culture media is unchanged by the presence of MSC microbeads, suggesting de novo sGAG synthesis in OA explants. Co-culture with MSC microbeads increased the DNA content and Ki67+ cells in OA explants, indicating proliferation. An increase in secreted cytokines IL-10, HGF, and sFAS assessed by multiplex cytokine assay, increased TIMP1 levels, and reduction in percent apoptotic cells in OA explants is noted. Together, data demonstrates that paracrine factors secreted by alginate encapsulated MSCs microbeads in response to OA cartilage, create an anabolic, proliferative, and anti-apoptotic microenvironment inducing endogenous regeneration in clinically relevant, patient-derived OA cartilage.
Collapse
Affiliation(s)
- Neety Sahu
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Pranay Agarwal
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Fiorella Grand
- Neurological Disease Institute, Gladstone Institutes, University of California, San Francisco, CA-94107
| | - Michela Bruschi
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Stuart Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Derek Ammanatullah
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| | - Nidhi Bhutani
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA-94305, USA
| |
Collapse
|
38
|
Cellhesion VP enhances the immunomodulating potential of human mesenchymal stem cell-derived extracellular vesicles. Biomaterials 2021; 271:120742. [PMID: 33706111 DOI: 10.1016/j.biomaterials.2021.120742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 02/17/2021] [Accepted: 02/25/2021] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cell (MSC) transplantation is a promising therapy for regenerative medicine. However, MSCs grown under two-dimensional (2D) culture conditions differ significantly in cell shape from those in the body, with downregulated stemness genes and secretion of paracrine factors. Here, we evaluated the effect of 3D culture using Cellhesion VP, a water-insoluble material composed of chitin-based polysaccharide fibers, on the characteristics of human Wharton's jelly-derived MSCs (hMSCs). Cellhesion VP significantly increased cell proliferation after retrieval. Transcriptome analyses suggested that genes involved in cell stemness, migration ability, and extracellular vesicle (EV) production were enhanced by 3D culture. Subsequent biochemical analyses showed that the expression levels of stemness genes including OCT4, NANOG, and SSEA4 were upregulated and migration capacity was elevated in 3D-cultured hMSCs. In addition, EV production was significantly elevated in 3D cells, which contained a distinct protein profile from 2D cells. Gene and drug connectivity analyses revealed that the 2D and 3D EVs had similar functions as immunomodulators; however, 3D EVs had completely distinct therapeutic profiles for various infectious and metabolic diseases based on activation of disease-associated signaling pathways. Therefore, EVs from Cellhesion VP-primed hMSCs offer a new treatment for immune and metabolic diseases.
Collapse
|
39
|
Sridharan R, Kelly DJ, O'Brien FJ. Substrate Stiffness Modulates the Crosstalk Between Mesenchymal Stem Cells and Macrophages. J Biomech Eng 2021; 143:031001. [PMID: 33067618 DOI: 10.1115/1.4048809] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Indexed: 07/25/2024]
Abstract
Upon implantation of a biomaterial, mesenchymal stem cells (MSCs) and macrophages contribute to the wound healing response and the regeneration cascade. Although biomaterial properties are known to direct MSC differentiation and macrophage polarization, the role of biomaterial cues, specifically stiffness, in directing the crosstalk between the two cell types is still poorly understood. This study aimed to elucidate the role of substrate stiffness in modulating the immunomodulatory properties of MSCs and to shed light on their complex interactions with macrophages when presented with diverse biomaterial stiffness cues, a situation analogous to the implant environment where multiple cell types interact with an implanted biomaterial to determine regenerative outcomes. We show that MSCs do not play an immunomodulatory role in the absence of an inflammatory stimulus. Using collagen-coated polyacrylamide gels of varying stiffness values, we demonstrate that the immunomodulatory capability of MSCs in the presence of an inflammatory stimulus is not dependent on the stiffness of the underlying substrate. Moreover, using paracrine and direct contact culture models, we show that a bidirectional crosstalk between MSCs and macrophages is necessary for promoting anti-inflammatory responses and positive immunomodulation, which is dependent on the stiffness of the underlying substrate. We finally show that direct cell-cell contact is not essential for this effect, with paracrine interactions promoting immunomodulatory interactions between MSCs and macrophages. Together, these results demonstrate that biophysical cues such as stiffness that are presented by biomaterials can be tuned to promote positive interactions between MSCs and macrophages which can in turn direct the downstream regenerative response.
Collapse
Affiliation(s)
- Rukmani Sridharan
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Daniel J Kelly
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2 D02 YN77, Ireland; Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland
| |
Collapse
|
40
|
Latest advances to enhance the therapeutic potential of mesenchymal stromal cells for the treatment of immune-mediated diseases. Drug Deliv Transl Res 2021; 11:498-514. [PMID: 33634433 DOI: 10.1007/s13346-021-00934-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cells (MSCs) present the capacity to secrete multiple immunomodulatory factors in response to their microenvironment. This property grants them a golden status among the novel alternatives to treat multiple diseases in which there is an unneeded or exaggerated immune response. However, important challenges still make difficult the clinical implementation of MSC-based therapies, being one of the most remarkable the lack of efficacy due to their transient immunomodulatory effects. To overcome this issue and boost the regulatory potential of MSCs, multiple strategies are currently being explored. Some of them consist of ex vivo pre-conditioning MSCs prior to their administration, including exposure to pro-inflammatory cytokines or to low oxygen concentrations. However, currently, alternative strategies that do not require such ex vivo manipulation are gaining special attention. Among them, the recreation of a three dimensional (3D) environment is remarkable. This approach has been reported to not only boost the immunomodulatory potential of MSCs but also increase their in vivo persistence and viability. The present work revises the therapeutic potential of MSCs, highlighting their immunomodulatory activity as a potential treatment for diseases caused by an exacerbated or unnecessary immune response. Moreover, it offers an updated vision of the most widely employed pre-conditioning strategies and 3D systems intended to enhance MSC-mediated immunomodulation, to conclude discussing the major challenges still to overcome in the field.
Collapse
|
41
|
Murata Y, Jo JI, Tabata Y. Visualization of Apoptosis in Three-Dimensional Cell Aggregates Based on Molecular Beacon Imaging. Tissue Eng Part C Methods 2021; 27:264-275. [PMID: 33619986 DOI: 10.1089/ten.tec.2020.0338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The objective of this study is to visualize cell apoptosis in three-dimensional (3D) cell aggregates based on molecular beacons (MB). Two types of MB for messenger RNA were used: caspase-3 MB as a target for apoptosis and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) MB as a control of stable fluorescence in cells. To enhance the MB internalization into cells, caspase-3 and GAPDH MB were incorporated in cationized gelatin nanospheres (cGNS), respectively (cGNScasp3 MB and cGNSGAP MB). In addition, cGNS co-incorporating caspase-3 and GAPDH MB (cGNSdual MB) were prepared to perform the dual-color imaging for the same cell aggregate. The cGNSMB were incubated with mouse mesenchymal stem cells to label with MB in the two-dimensional culture. The cell apoptosis mediated by the addition of antibody for a death receptor Fas was ratiometrically detected by the cGNSdual MB to the same extent as single MB. The cell aggregates were prepared from MB-labeled cells, and the MB fluorescence was detected from almost all the cells even in the 3D aggregates to show the homogenous distribution. In addition to the Fas-mediated apoptosis, the aggregates were treated with camptothecin of a low-molecular weight apoptosis inducer. The fluorescence of caspase-3 MB was mainly distributed at the surface surrounding site of Fas-mediated apoptotic aggregates rather than the center site, while that of GAPDH MB was detected even in the interior site. On the other hand, in the camptothecin-induced apoptotic aggregates, both caspae-3 and GAPDH MB fluorescence were detected from the interior site of aggregates as well as the surrounding site. It is likely that the MB fluorescence reflected the localization of apoptotic position caused by the different molecular sizes of apoptosis inducer and the consequent penetration into the aggregates. It is concluded that the cGMSMB are a promising system to visualize cell apoptosis in 3D cell aggregates without the destruction of aggregates.
Collapse
Affiliation(s)
- Yuki Murata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
42
|
Schmitz C, Potekhina E, Belousov VV, Lavrentieva A. Hypoxia Onset in Mesenchymal Stem Cell Spheroids: Monitoring With Hypoxia Reporter Cells. Front Bioeng Biotechnol 2021; 9:611837. [PMID: 33614611 PMCID: PMC7892969 DOI: 10.3389/fbioe.2021.611837] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
The therapeutic and differentiation potential of human mesenchymal stems cells (hMSCs) makes these cells a promising candidate for cellular therapies and tissue engineering. On the path of a successful medical application of hMSC, the cultivation of cells in a three-dimensional (3D) environment was a landmark for the transition from simple two-dimensional (2D) testing platforms to complex systems that mimic physiological in vivo conditions and can improve hMSC curative potential as well as survival after implantation. A 3D arrangement of cells can be mediated by scaffold materials where cells get entrapped in pores, or by the fabrication of spheroids, scaffold-free self-organized cell aggregates that express their own extracellular matrix. Independently from the cultivation method, cells expanded in 3D experience an inhomogeneous microenvironment. Many gradients in nutrient supply, oxygen supply, and waste disposal from one hand mimic in vivo microenvironment, but also put every cell in the 3D construct in a different context. Since oxygen concentration in spheroids is compromised in a size-dependent manner, it is crucial to have a closer insight on the thresholds of hypoxic response in such systems. In this work, we want to improve our understanding of oxygen availability and consequensing hypoxia onset in hMSC spheroids. Therefore, we utilized human adipose tissue-derived MSCs (hAD-MSCs) modified with a genetical sensor construct to reveal (I) the influence of spheroid production methods and (II) hMSCs cell number per spheroid to detect the onset of hypoxia in aggregates. We could demonstrate that not only higher cell numbers of MSCs, but also spheroid formation method plays a critical role in onset of hypoxia.
Collapse
Affiliation(s)
- Carola Schmitz
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| | - Ekaterina Potekhina
- Department of Metabolism and Redox Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies, Federal Biomedical Agency, Moscow, Russia
| | - Antonina Lavrentieva
- Institute of Technical Chemistry, Gottfried Wilhelm Leibniz University Hannover, Hanover, Germany
| |
Collapse
|
43
|
Bhandary L, Bailey PC, Chang KT, Underwood KF, Lee CJ, Whipple RA, Jewell CM, Ory E, Thompson KN, Ju JA, Mathias TM, Pratt SJP, Vitolo MI, Martin SS. Lipid tethering of breast tumor cells reduces cell aggregation during mammosphere formation. Sci Rep 2021; 11:3214. [PMID: 33547369 PMCID: PMC7865010 DOI: 10.1038/s41598-021-81919-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Mammosphere assays are widely used in vitro to identify prospective cancer-initiating stem cells that can propagate clonally to form spheres in free-floating conditions. However, the traditional mammosphere assay inevitably introduces cell aggregation that interferes with the measurement of true mammosphere forming efficiency. We developed a method to reduce tumor cell aggregation and increase the probability that the observed mammospheres formed are clonal in origin. Tethering individual tumor cells to lipid anchors prevents cell drift while maintaining free-floating characteristics. This enables real-time monitoring of single tumor cells as they divide to form mammospheres. Monitoring tethered breast cancer cells provided detailed size information that correlates directly to previously published single cell tracking data. We observed that 71% of the Day 7 spheres in lipid-coated wells were between 50 and 150 μm compared to only 37% in traditional low attachment plates. When an equal mixture of MCF7-GFP and MCF7-mCherry cells were seeded, 65% of the mammospheres in lipid-coated wells demonstrated single color expression whereas only 32% were single-colored in low attachment wells. These results indicate that using lipid tethering for mammosphere growth assays can reduce the confounding factor of cell aggregation and increase the formation of clonal mammospheres.
Collapse
Affiliation(s)
- Lekhana Bhandary
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Patrick C Bailey
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA.,Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Katarina T Chang
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA.,Graduate Program in Life Sciences, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Karen F Underwood
- UMGCCC Flow Cytometry Shared Service, 655 West Baltimore Street, BRB 7-022, Baltimore, MD, 21201, USA
| | - Cornell J Lee
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Rebecca A Whipple
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, 3102 A. James Clark Hall, College Park, MD, 20742, USA
| | - Eleanor Ory
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Keyata N Thompson
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Julia A Ju
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Trevor M Mathias
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA
| | - Stephen J P Pratt
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA.,Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Michele I Vitolo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA. .,Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD, 21201, USA. .,Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| | - Stuart S Martin
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine (UMGCCC), 22 S. Greene St., Baltimore, MD, 21201, USA. .,Graduate Program in Biochemistry, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD, 21201, USA. .,Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA. .,, Bressler Research Building Room 10-29, 655 West Baltimore Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
44
|
Pouraghaei Sevari S, Ansari S, Chen C, Moshaverinia A. Harnessing Dental Stem Cell Immunoregulation Using Cell-Laden Biomaterials. J Dent Res 2021; 100:568-575. [PMID: 33478322 DOI: 10.1177/0022034520985820] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Successful tissue engineering therapies rely on the appropriate selection of the cell source, biomaterial, and regulatory factors. To be applied in a wide range of clinical applications, the ideal cell source needs to be easily accessible and abundant. Human orofacial tissues and teeth harbor several populations of mesenchymal stem cells (MSCs) with self-renewal and multilineage differentiation capabilities. The ease of access, relative abundance, and minimally invasive isolation procedures needed to harvest most types of the dental-derived MSCs render them a promising cell source for tissue engineering applications. A growing body of evidence has reported the profound immunoregulatory potential of dental-derived MSCs as compared with their bone marrow counterparts. Biomaterials can act as a physical barrier protecting the MSCs from the invasion of the immune system by hindering penetration of proinflammatory cells/cytokines, leading to higher viability of the encapsulated MSCs and improved tissue regeneration. Besides their protective capabilities, biomaterials can actively contribute to the immunoregulatory potential of the MSCs through their physical and chemical properties, including porosity and elasticity. However, despite recent advancement, the therapeutic capability of biomaterials to regulate the MSC-host immune system crosstalk and the mechanism underlying this immunoregulation has been poorly understood. It has been reported that biomaterials can regulate the viability and determine the fate of the encapsulated MSCs through modulation of the NF-kB pathway and the caspase-3 and caspase-8 proapoptotic cascades. Additionally, the physiomechanical properties of the encapsulating biomaterial have been shown to modulate clustering of TNF-α receptors on the encapsulated MSCs while regulating the production of anti-inflammatory factors such as indoleamine 2,3-dioxygenase (IDO) and prostaglandin E2 (PGE2) through activation of the P38 MAPK pathway. In the current review, we sought to provide a thorough overview of the immunomodulatory functions of dental-derived MSCs and the role of biomaterials in their interplay with the host immune system.
Collapse
Affiliation(s)
- S Pouraghaei Sevari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - S Ansari
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - C Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Center of Innovation and Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - A Moshaverinia
- Weintraub Center for Reconstructive Biotechnology, Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
45
|
Miceli V, Bulati M, Iannolo G, Zito G, Gallo A, Conaldi PG. Therapeutic Properties of Mesenchymal Stromal/Stem Cells: The Need of Cell Priming for Cell-Free Therapies in Regenerative Medicine. Int J Mol Sci 2021; 22:763. [PMID: 33466583 PMCID: PMC7828743 DOI: 10.3390/ijms22020763] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) are multipotent adult stem cells that support homeostasis during tissue regeneration. In the last decade, cell therapies based on the use of MSCs have emerged as a promising strategy in the field of regenerative medicine. Although these cells possess robust therapeutic properties that can be applied in the treatment of different diseases, variables in preclinical and clinical trials lead to inconsistent outcomes. MSC therapeutic effects result from the secretion of bioactive molecules affected by either local microenvironment or MSC culture conditions. Hence, MSC paracrine action is currently being explored in several clinical settings either using a conditioned medium (CM) or MSC-derived exosomes (EXOs), where these products modulate tissue responses in different types of injuries. In this scenario, MSC paracrine mechanisms provide a promising framework for enhancing MSC therapeutic benefits, where the composition of secretome can be modulated by priming of the MSCs. In this review, we examine the literature on the priming of MSCs as a tool to enhance their therapeutic properties applicable to the main processes involved in tissue regeneration, including the reduction of fibrosis, the immunomodulation, the stimulation of angiogenesis, and the stimulation of resident progenitor cells, thereby providing new insights for the therapeutic use of MSCs-derived products.
Collapse
Affiliation(s)
- Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), 90127 Palermo, Italy; (M.B.); (G.I.); (G.Z.); (A.G.); (P.G.C.)
| | | | | | | | | | | |
Collapse
|
46
|
Krutty JD, Koesser K, Schwartz S, Yun J, Murphy WL, Gopalan P. Xeno-Free Bioreactor Culture of Human Mesenchymal Stromal Cells on Chemically Defined Microcarriers. ACS Biomater Sci Eng 2021; 7:617-625. [PMID: 33448784 DOI: 10.1021/acsbiomaterials.0c00663] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Human mesenchymal stromal cells (hMSC), also called mesenchymal stem cells, are adult cells that have demonstrated their potential in therapeutic applications, highlighted by their ability to differentiate down different lineages, modulate the immune system, and produce biologics. There is a pressing need for scalable culture systems for hMSC due to the large number of cells needed for clinical applications. Most current methods for expanding hMSC fail to provide a reproducible cell product in clinically required cell numbers without the use of serum-containing media or harsh enzymes. In this work, we apply a tailorable, thin, synthetic polymer coating-poly(poly(ethylene glycol) methyl ether methacrylate-ran-vinyl dimethyl azlactone-ran-glycidyl methacrylate) (P(PEGMEMA-r-VDM-r-GMA), PVG)-to the surface of commercially available polystyrene (PS) microcarriers to create chemically defined three-dimensional (3D) surfaces for large-scale cell expansion. These chemically defined microcarriers provide a reproducible surface that does not rely on the adsorption of xenogeneic serum proteins to mediate cell adhesion, enabling their use in xeno-free culture systems. Specifically, this work demonstrates the improved adhesion of hMSC to coated microcarriers over PS microcarriers in xeno-free media and describes their use in a readily scalable, bioreactor-based culture system. Additionally, these surfaces resist the adsorption of media-borne and cell-produced proteins, which result in integrin-mediated cell adhesion throughout the culture period. This feature allows the cells to be efficiently passaged from the microcarrier using a chemical chelating agent (ethylenediaminetetraacetic acid (EDTA)) in the absence of cleavage enzymes, an improvement over other microcarrier products in the field. Bioreactor culture of hMSC on these microcarriers enabled the production of hMSC over 4 days from a scalable, xeno-free environment.
Collapse
Affiliation(s)
- John D Krutty
- Department of Materials Science and Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| | - Kevin Koesser
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| | - Stephen Schwartz
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| | - Junsu Yun
- Department of Materials Science and Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| | - William L Murphy
- Department of Materials Science and Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States.,Department of Biomedical Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States.,Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| | - Padma Gopalan
- Department of Materials Science and Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States.,Department of Biomedical Engineering, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States.,Department of Chemistry, University of Wisconsin-Madison, 1500 University Avenue, Madison, Wisconsin 53706, United States
| |
Collapse
|
47
|
Bicer M, Cottrell GS, Widera D. Impact of 3D cell culture on bone regeneration potential of mesenchymal stromal cells. Stem Cell Res Ther 2021; 12:31. [PMID: 33413646 PMCID: PMC7791873 DOI: 10.1186/s13287-020-02094-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
As populations age across the world, osteoporosis and osteoporosis-related fractures are becoming the most prevalent degenerative bone diseases. More than 75 million patients suffer from osteoporosis in the USA, the EU and Japan. Furthermore, it is anticipated that the number of patients affected by osteoporosis will increase by a third by 2050. Although conventional therapies including bisphosphonates, calcitonin and oestrogen-like drugs can be used to treat degenerative diseases of the bone, they are often associated with serious side effects including the development of oesophageal cancer, ocular inflammation, severe musculoskeletal pain and osteonecrosis of the jaw.The use of autologous mesenchymal stromal cells/mesenchymal stem cells (MSCs) is a possible alternative therapeutic approach to tackle osteoporosis while overcoming the limitations of traditional treatment options. However, osteoporosis can cause a decrease in the numbers of MSCs, induce their senescence and lower their osteogenic differentiation potential.Three-dimensional (3D) cell culture is an emerging technology that allows a more physiological expansion and differentiation of stem cells compared to cultivation on conventional flat systems.This review will discuss current understanding of the effects of different 3D cell culture systems on proliferation, viability and osteogenic differentiation, as well as on the immunomodulatory and anti-inflammatory potential of MSCs.
Collapse
Affiliation(s)
- Mesude Bicer
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK
| | - Graeme S Cottrell
- Cellular and Molecular Neuroscience, School of Pharmacy, University of Reading, Reading, UK
| | - Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, Reading School of Pharmacy, University of Reading, PO Box 226, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
48
|
Zhu M, Hua T, Ouyang T, Qian H, Yu B. Applications of Mesenchymal Stem Cells in Liver Fibrosis: Novel Strategies, Mechanisms, and Clinical Practice. Stem Cells Int 2021; 2021:6546780. [PMID: 34434239 PMCID: PMC8380491 DOI: 10.1155/2021/6546780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/07/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Liver fibrosis is a common result of most chronic liver diseases, and advanced fibrosis often leads to cirrhosis. Currently, there is no effective treatment for liver cirrhosis except liver transplantation. Therefore, it is important to carry out antifibrosis treatment to reverse liver damage in the early stage of liver fibrosis. Mesenchymal stem cells (MSCs) are the most widely used stem cells in the field of regenerative medicine. The preclinical and clinical research results of MSCs in the treatment of liver fibrosis and cirrhosis show that MSC administration is a promising treatment for liver fibrosis and cirrhosis. MSCs reverse liver fibrosis and increase liver function mainly through differentiation into hepatocytes, immune regulation, secretion of cytokines and other nutritional factors, reduction of hepatocyte apoptosis, and promotion of hepatocyte regeneration. Recently, many studies provided a variety of new methods and strategies to improve the effect of MSCs in the treatment of liver fibrosis. In this review, we summarized the current effective methods and strategies and their potential mechanisms of MSCs in the treatment of liver fibrosis, as well as the current research progress in clinical practice. We expect to achieve complete reversal of liver injury with MSC-based therapy in the future.
Collapse
Affiliation(s)
- Mengmei Zhu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Tianzhen Hua
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Tao Ouyang
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Huofu Qian
- 2Department of Gastroenterology, The Second People's Hospital of Taizhou, China
| | - Bing Yu
- 1Department of Cell Biology, Center for Stem Cell and Medicine, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| |
Collapse
|
49
|
Gorgun C, Ceresa D, Lesage R, Villa F, Reverberi D, Balbi C, Santamaria S, Cortese K, Malatesta P, Geris L, Quarto R, Tasso R. Dissecting the effects of preconditioning with inflammatory cytokines and hypoxia on the angiogenic potential of mesenchymal stromal cell (MSC)-derived soluble proteins and extracellular vesicles (EVs). Biomaterials 2020; 269:120633. [PMID: 33453634 DOI: 10.1016/j.biomaterials.2020.120633] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023]
Abstract
Mesenchymal stromal cells (MSCs) are characterized by a regulatory phenotype and respond promptly to the environmental signals modulating their secretory activity. An appropriate preconditioning may induce MSCs to release secretomes with an enhanced regenerative potential. However, it fails to take into account that secretomes are composed by both soluble factors and extracellular vesicles (EVs), whose functions could be altered differently by the preconditioning approach. Here we demonstrate that the MSC secretome is strongly modulated by the simultaneous stimulation with hypoxia and pro-inflammatory cytokines, used to mimic the harsh environment present at the site of injury. We observed that the environmental variations strongly influenced the angiogenic potential of the different secretome fractions. Upon inflammation, the pro-angiogenic capacity of the soluble component of the MSC secretome was strongly inhibited, regardless of the oxygen level, while the EV-encapsulated component was not significantly affected by the inflammatory stimuli. These effects were accompanied by the modulation of the secreted proteins. On one hand, inflammation-activated MSCs release proteins mainly involved in the interaction with innate immune cells and in tissue remodeling/repair; on the other hand, when MSCs are not exposed to an inflamed environment, they respond to the different oxygen levels modulating the expression of proteins involved in the angiogenic process. The cargo content (in terms of miRNAs) of the corresponding EV fractions was less sensitive to the influence of the external stimuli. Our findings suggest that the therapeutic efficacy of MSC-based therapies could be enhanced by selecting the appropriate preconditioning approach and carefully discriminating its effects on the different secretome components.
Collapse
Affiliation(s)
- Cansu Gorgun
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy; U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Davide Ceresa
- U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Raphaelle Lesage
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium; Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Federico Villa
- U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniele Reverberi
- U.O. Molecular Pathology, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Foundation, 6900, Lugano, Switzerland
| | - Sara Santamaria
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Katia Cortese
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Paolo Malatesta
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy; U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Liesbet Geris
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium; Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium; Biomechanics Research Unit, GIGA in Silico Medicine, University of Liège, Liège, Belgium
| | - Rodolfo Quarto
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy; U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Roberta Tasso
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy; U.O. Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
50
|
Burnham AJ, Foppiani EM, Horwitz EM. Key Metabolic Pathways in MSC-Mediated Immunomodulation: Implications for the Prophylaxis and Treatment of Graft Versus Host Disease. Front Immunol 2020; 11:609277. [PMID: 33365034 PMCID: PMC7750397 DOI: 10.3389/fimmu.2020.609277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are spindle-shaped, plastic-adherent cells in vitro with potent immunosuppressive activity both in vitro and in vivo. MSCs have been employed as a cellular immunotherapy in diverse preclinical models and clinical trials, but most commonly as agents for the prophylaxis or therapy of graft versus host disease after hematopoietic cell transplantation. In addition to the oft studied secreted cytokines, several metabolic pathways intrinsic to MSCs, notably indoleamine 2,3-dioxygenase, prostaglandin E2, hypoxia-inducible factor 1 α, heme oxygenase-1, as well as energy-generating metabolism, have been shown to play roles in the immunomodulatory activity of MSCs. In this review, we discuss these key metabolic pathways in MSCs which have been reported to contribute to MSC therapeutic effects in the setting of hematopoietic cell transplantation and graft versus host disease. Understanding the contribution of MSC metabolism to immunomodulatory activity may substantially inform the development of future clinical applications of MSCs.
Collapse
Affiliation(s)
- Andre J Burnham
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Elisabetta Manuela Foppiani
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Edwin M Horwitz
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, United States.,Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|