151
|
Saadh MJ, Ramírez-Coronel AA, Saini RS, Arias-Gonzáles JL, Amin AH, Gavilán JCO, Sârbu I. Advances in mesenchymal stem/stromal cell-based therapy and their extracellular vesicles for skin wound healing. Hum Cell 2023:10.1007/s13577-023-00904-8. [PMID: 37067766 DOI: 10.1007/s13577-023-00904-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023]
Abstract
Wound healing is a dynamic and complicated process containing overlapping phases. Presently, definitive therapy is not available, and the investigation into optimal wound care is influenced by the efficacy and cost-effectiveness of developing therapies. Accumulating evidence demonstrated the potential role of mesenchymal stem/stromal cell (MSC) therapy in several tissue injuries and diseases due to their high proliferation and differentiation abilities along with an easy collection procedure, low tumorigenesis, and immuno-privileged status. MSCs have also accelerated wound repair in all phases through their advantageous properties, such as accelerating wound closure, improving re-epithelialization, elevating angiogenesis, suppressing inflammation, and modulating extracellular matrix (ECM) remodeling. In addition, the beneficial therapeutic impacts of MSCs are largely associated with their paracrine functions, including extracellular vesicles (EVs). Exosomes and microvesicles are the two main subgroups of EVs. These vesicles are heterogeneous bilayer membrane structures that contain several proteins, lipids, and nucleic acids. EVs have emerged as a promising alternative to stem cell-based therapies because of their lower immunogenicity, tumorigenicity, and ease of management. MSCs from various sources have been widely investigated in skin wound healing and regeneration. Considering these features, in this review, we highlighted recent studies that the investigated therapeutic potential of various MSCs and MSC-EVs in skin damages and wounds.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellín, Colombia
| | | | - José Luis Arias-Gonzáles
- Department of Social Sciences, Faculty of Social Studies, Pontifical University of Peru, San Miguel, Peru
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | | | - Ioan Sârbu
- 2nd Department of Surgery, Pediatric Surgery and Orthopedics, "Grigore T. Popa", University of Medicine and Pharmacy, 700115, Iași, Romania.
| |
Collapse
|
152
|
Tung S, Delavogia E, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S. Harnessing the therapeutic potential of the stem cell secretome in neonatal diseases. Semin Perinatol 2023; 47:151730. [PMID: 36990921 PMCID: PMC10133192 DOI: 10.1016/j.semperi.2023.151730] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Preterm birth and intrapartum related complications account for a substantial amount of mortality and morbidity in the neonatal period despite significant advancements in neonatal-perinatal care. Currently, there is a noticeable lack of curative or preventative therapies available for any of the most common complications of prematurity including bronchopulmonary dysplasia, necrotizing enterocolitis, intraventricular hemorrhage, periventricular leukomalacia and retinopathy of prematurity or hypoxic-ischemic encephalopathy, the main cause of perinatal brain injury in term infants. Mesenchymal stem/stromal cell-derived therapy has been an active area of investigation for the past decade and has demonstrated encouraging results in multiple experimental models of neonatal disease. It is now widely acknowledged that mesenchymal stem/stromal cells exert their therapeutic effects via their secretome, with the principal vector identified as extracellular vesicles. This review will focus on summarizing the current literature and investigations on mesenchymal stem/stromal cell-derived extracellular vesicles as a treatment for neonatal diseases and examine the considerations to their application in the clinical setting.
Collapse
Affiliation(s)
- Stephanie Tung
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Eleni Delavogia
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States; Department of Pediatrics, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
153
|
Thébaud B. Stem cell therapies for neonatal lung diseases: Are we there yet? Semin Perinatol 2023; 47:151724. [PMID: 36967368 DOI: 10.1016/j.semperi.2023.151724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Lung diseases are a main cause of mortality and morbidity in neonates. Despite major breakthroughs, therapies remain supportive and, in some instances, contribute to lung injury. Because the neonatal lung is still developing, the ideal therapy should be capable of preventing/repairing lung injury while at the same time, promoting lung growth. Cell-based therapies hold high hopes based on laboratory experiments in animal models of neonatal lung injury. Mesenchymal stromal cells and amnion epithelial cells are now in early phase clinical trials to test the feasibility, safety and early signs of efficacy in preterm infants at risk of developing bronchopulmonary dysplasia. Other cell-based therapies are being explored in experimental models of congenital diaphragmatic hernia and alveolar capillary dysplasia. This review will summarize current evidence that has lead to the clinical translation of cell-based therapies and highlights controversies and the numerous questions that remain to be addressed to harness the putative repair potential of cell-based therapies.
Collapse
Affiliation(s)
- Bernard Thébaud
- Regenerative Medicine Program, The Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada.; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.; Neonatology, Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
154
|
Wieder R. Fibroblasts as Turned Agents in Cancer Progression. Cancers (Basel) 2023; 15:2014. [PMID: 37046676 PMCID: PMC10093070 DOI: 10.3390/cancers15072014] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Differentiated epithelial cells reside in the homeostatic microenvironment of the native organ stroma. The stroma supports their normal function, their G0 differentiated state, and their expansion/contraction through the various stages of the life cycle and physiologic functions of the host. When malignant transformation begins, the microenvironment tries to suppress and eliminate the transformed cells, while cancer cells, in turn, try to resist these suppressive efforts. The tumor microenvironment encompasses a large variety of cell types recruited by the tumor to perform different functions, among which fibroblasts are the most abundant. The dynamics of the mutual relationship change as the sides undertake an epic battle for control of the other. In the process, the cancer "wounds" the microenvironment through a variety of mechanisms and attracts distant mesenchymal stem cells to change their function from one attempting to suppress the cancer, to one that supports its growth, survival, and metastasis. Analogous reciprocal interactions occur as well between disseminated cancer cells and the metastatic microenvironment, where the microenvironment attempts to eliminate cancer cells or suppress their proliferation. However, the altered microenvironmental cells acquire novel characteristics that support malignant progression. Investigations have attempted to use these traits as targets of novel therapeutic approaches.
Collapse
Affiliation(s)
- Robert Wieder
- Rutgers New Jersey Medical School and the Cancer Institute of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
155
|
Song WP, Jin LY, Zhu MD, Wang H, Xia DS. Clinical trials using dental stem cells: 2022 update. World J Stem Cells 2023; 15:31-51. [PMID: 37007456 PMCID: PMC10052340 DOI: 10.4252/wjsc.v15.i3.31] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/20/2023] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
For nearly 20 years, dental stem cells (DSCs) have been successfully isolated from mature/immature teeth and surrounding tissue, including dental pulp of permanent teeth and exfoliated deciduous teeth, periodontal ligaments, dental follicles, and gingival and apical papilla. They have several properties (such as self-renewal, multidirectional differentiation, and immunomodulation) and exhibit enormous potential for clinical applications. To date, many clinical articles and clinical trials using DSCs have reported the treatment of pulpitis, periapical lesions, periodontitis, cleft lip and palate, acute ischemic stroke, and so on, and DSC-based therapies obtained satisfactory effects in most clinical trials. In these studies, no adverse events were reported, which suggested the safety of DSC-based therapy. In this review, we outline the characteristics of DSCs and summarize clinical trials and their safety as DSC-based therapies. Meanwhile, we also present the current limitations and perspectives of DSC-based therapy (such as harvesting DSCs from inflamed tissue, applying DSC-conditioned medium/DSC-derived extracellular vesicles, and expanding-free strategies) to provide a theoretical basis for their clinical applications.
Collapse
Affiliation(s)
- Wen-Peng Song
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Lu-Yuan Jin
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Meng-Di Zhu
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China
| | - Hao Wang
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Deng-Sheng Xia
- Department of General Dentistry and Integrated Emergency Dental Care, Beijing Stomatological Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
156
|
Emam M, Cohen C, Willeford S, Mahesh K, Le MQ, Wilckens J. Role of Conservative Treatment vs Surgical Treatment for Rotator Cuff Tears: A Narrative Review. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2023. [DOI: 10.1007/s40141-023-00389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
|
157
|
Obesity and Wound Healing: Focus on Mesenchymal Stem Cells. Life (Basel) 2023; 13:life13030717. [PMID: 36983872 PMCID: PMC10059997 DOI: 10.3390/life13030717] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/20/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Chronic wounds represent nowadays a major challenge for both clinicians and researchers in the regenerative setting. Obesity represents one of the major comorbidities in patients affected by chronic ulcers and therefore diverse studies aimed at assessing possible links between these two morbid conditions are currently ongoing. In particular, adipose tissue has recently been described as having metabolic and endocrine functions rather than serving as a mere fat storage deposit. In this setting, adipose-derived stem cells, a peculiar subset of mesenchymal stromal/stem cells (MSCs) located in adipose tissue, have been demonstrated to possess regenerative and immunological functions with a key role in regulating both adipocyte function and skin regeneration. The aim of the present review is to give an overview of the most recent findings on wound healing, with a special focus on adipose tissue biology and obesity.
Collapse
|
158
|
Pinkhasov I, Kabakov L, Nemcovsky CE, Weinreb M, Schlesinger P, Bender O, Gal M, Bar DZ, Weinberg E. Single-cell transcriptomic analysis of oral masticatory and lining mucosa-derived mesenchymal stromal cells. J Clin Periodontol 2023; 50:807-818. [PMID: 36864739 DOI: 10.1111/jcpe.13799] [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: 11/03/2022] [Revised: 01/25/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
AIM To reveal the heterogeneity of ex vivo-cultured human mesenchymal stromal cells derived from either masticatory or lining oral mucosa. MATERIALS AND METHODS Cells were retrieved from the lamina propria of the hard palate and alveolar mucosa of three individuals. The analysis of transcriptomic-level differences was accomplished using single-cell RNA sequencing. RESULTS Cluster analysis clearly distinguished between cells from the masticatory and lining oral mucosa, and revealed 11 distinct cell sub-populations, annotated as fibroblasts, smooth muscle cells or mesenchymal stem cells. Interestingly, cells presenting a mesenchymal stem cell-like gene expression pattern were predominantly found in masticatory mucosa. Although cells of masticatory mucosa origin were highly enriched for biological processes associated with wound healing, those from the lining oral mucosa were highly enriched for biological processes associated with the regulation of epithelial cells. CONCLUSIONS Our previous work had shown that cells from the lining and masticatory oral mucosae are phenotypically heterogeneous. Here, we extend these findings to show that these changes are not the result of differences in averages but rather represent two distinct cell populations, with mesenchymal stem cells more common in masticatory mucosa. These features may contribute to specific physiological functions and have relevance for potential therapeutic interventions.
Collapse
Affiliation(s)
- Ilan Pinkhasov
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liron Kabakov
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Carlos E Nemcovsky
- Department of Periodontology and Oral Implantology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Miron Weinreb
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pnina Schlesinger
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maayan Gal
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Evgeny Weinberg
- Department of Oral Biology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Periodontology and Oral Implantology, Goldschleger School of Dental Medicine, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
159
|
Ciccocioppo R, Guadalajara H, Astori G, Carlino G, García-Olmo D. Misconceptions, hurdles and recommendations regarding the use of mesenchymal stem/stromal cells in perianal Crohn disease. Cytotherapy 2023; 25:230-234. [PMID: 36543715 DOI: 10.1016/j.jcyt.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/14/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy.
| | - Hector Guadalajara
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| | - Giuseppe Astori
- Laboratory of Advanced Cellular Therapies, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Giorgio Carlino
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy
| | - Damián García-Olmo
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| |
Collapse
|
160
|
Frias F, Matos B, Jarnalo M, Freitas-Ribeiro S, Reis RL, Pirraco RP, Horta R. Stromal Vascular Fraction Obtained From Subcutaneous Adipose Tissue: Ex-Obese and Older Population as Main Clinical Targets. J Surg Res 2023; 283:632-639. [PMID: 36446251 DOI: 10.1016/j.jss.2022.11.012] [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: 01/05/2022] [Revised: 10/12/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Human adipose tissue contains a heterogeneous and synergistic mixture of cells called stromal vascular fraction (SVF) with highly proliferative and angiogenic properties, conferring promising applicability in the field of regenerative medicine. This study aims to investigate if age, body mass index (BMI), history of obesity and massive weight loss, and harvest site are related to SVF cell marker expression. METHODS A total of 26 samples of subcutaneous adipose tissue were harvested from patients admitted to the Plastic and Reconstructive department in University Hospital Center of São João, Porto, Portugal, for body contouring surgery. The percentage of cells expressing CD31, CD34, CD45, CD73, CD90, and CD105 was assessed and compared with patient's age, BMI, history of obesity and massive weight loss (ex-obese group), and harvest site. RESULTS In the ex-obese group, a significantly higher number of cells expressing CD90 (P = 0.002) was found. BMI, harvest site, and age appear to have no association with SVF subpopulations. CONCLUSIONS This study suggests that ex-obese patients have a higher percentage of SVF cells expressing CD90, which correlates with higher proliferative and angiogenic rates. The effect of former obesity and massive weight loss on the expression of CD90 is a new and relevant finding because it makes this population a suitable candidate for reconstructive and aesthetic surgery and other fields of regenerative medicine. The use of SVF appears also promising in older patients because no negative correlation between increasing age and different cell markers expression was found.
Collapse
Affiliation(s)
- Francisca Frias
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal.
| | - Beatriz Matos
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| | - Mariana Jarnalo
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| | - Sara Freitas-Ribeiro
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Ricardo Horta
- Department of Plastic, Reconstructive and Maxillo-Facial Surgery, and Burn Unit, Centro Hospitalar de São João, University of Porto, Porto, Portugal
| |
Collapse
|
161
|
Kirkham AM, Bailey AJM, Shorr R, Lalu MM, Fergusson DA, Allan DS. Systematic review and meta-analysis of randomized controlled trials of mesenchymal stromal cells to treat coronavirus disease 2019: is it too late? Cytotherapy 2023; 25:341-352. [PMID: 36333234 PMCID: PMC9556962 DOI: 10.1016/j.jcyt.2022.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/08/2022] [Accepted: 10/05/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AIMS Evidence regarding the extent that mesenchymal stromal cells (MSCs) may improve clinical outcomes in patients with coronavirus disease 2019 (COVID-19) has been limited by marked inter-study heterogeneity, inconsistent product characterization and appreciable risk of bias (RoB). Given the evolution of treatment options and trajectory of the pandemic, an updated analysis of high-quality evidence from randomized controlled trials is needed for a timely and conclusive understanding of the effectiveness of MSCs. METHODS A systematic literature search through March 30, 2022, identified all English language, full-text randomized controlled trials examining the use of MSCs in the treatment of COVID-19. RESULTS Eight studies were identified (316 patients, 165 administered MSCs and 151 controls). Controls evolved significantly over time with a broad range of comparison treatments. All studies reported mortality at study endpoint. Random effects meta-analysis revealed that MSCs decreased relative risk of death (risk ratio, 0.63, 95% confidence interval, 0.42-0.94, P = 0.02, I2 = 14%) with no significant difference in absolute risk of death. MSCs decreased length of hospital stay and C-reactive protein levels and increased odds of clinical improvement at study endpoint compared with controls. Rates of adverse events and severe adverse events were similar between MSC and control groups. Only two (25%) studies reported all four International Society for Cell & Gene Therapy criteria for MSC characterization. Included studies had low (n = 7) or some (n = 1) concerns regarding RoB. CONCLUSIONS MSCs may reduce risk of death in patients with severe or critical COVID-19 and improve secondary clinical outcomes. Variable outcome reporting, inconsistent product characterization and variable control group treatments remain barriers to higher-quality evidence and may constrain clinical usage. A master protocol is proposed and appears necessary for accelerated translation of higher-quality evidence for future applications of MSC therapy.
Collapse
Affiliation(s)
- Aidan M Kirkham
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Adrian J M Bailey
- Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Risa Shorr
- Medical Information and Learning Services, The Ottawa Hospital, Ottawa, Canada
| | - Manoj M Lalu
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada; Department of Anesthesiology and Pain Medicine, University of Ottawa, Ottawa, Canada; Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada; Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Anesthesia, The Ottawa Hospital, Ottawa, Canada
| | - Dean A Fergusson
- Department of Medicine, University of Ottawa, Ottawa, Canada; Department of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Canada; Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - David S Allan
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada; Department of Medicine, University of Ottawa, Ottawa, Canada; Clinical Epidemiology, Ottawa Hospital Research Institute, Ottawa, Canada; Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Canada; Department of Medicine, The Ottawa Hospital, Ottawa, Canada.
| |
Collapse
|
162
|
Extracellular Vesicles and MicroRNA in Myelodysplastic Syndromes. Cells 2023; 12:cells12040658. [PMID: 36831325 PMCID: PMC9955013 DOI: 10.3390/cells12040658] [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: 12/26/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The bone marrow niche plays an increasing role in the pathophysiogenesis of myelodysplastic syndromes. More specifically, mesenchymal stromal cells, which can secrete extracellular vesicles and their miRNA contents, modulate the fate of hematopoietic stem cells leading to leukemogenesis. Extracellular vesicles can mediate their miRNA and protein contents between nearby cells but also in the plasma of the patients, being potent tools for diagnosis and prognostic markers in MDS. They can be targeted by antisense miRNA or by modulators of the secretion of extracellular vesicles and could lead to future therapeutic directions in MDS.
Collapse
|
163
|
Beleño Acosta B, Advincula RC, Grande-Tovar CD. Chitosan-Based Scaffolds for the Treatment of Myocardial Infarction: A Systematic Review. Molecules 2023; 28:1920. [PMID: 36838907 PMCID: PMC9962426 DOI: 10.3390/molecules28041920] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Cardiovascular diseases (CVD), such as myocardial infarction (MI), constitute one of the world's leading causes of annual deaths. This cardiomyopathy generates a tissue scar with poor anatomical properties and cell necrosis that can lead to heart failure. Necrotic tissue repair is required through pharmaceutical or surgical treatments to avoid such loss, which has associated adverse collateral effects. However, to recover the infarcted myocardial tissue, biopolymer-based scaffolds are used as safer alternative treatments with fewer side effects due to their biocompatibility, chemical adaptability and biodegradability. For this reason, a systematic review of the literature from the last five years on the production and application of chitosan scaffolds for the reconstructive engineering of myocardial tissue was carried out. Seventy-five records were included for review using the "preferred reporting items for systematic reviews and meta-analyses" data collection strategy. It was observed that the chitosan scaffolds have a remarkable capacity for restoring the essential functions of the heart through the mimicry of its physiological environment and with a controlled porosity that allows for the exchange of nutrients, the improvement of the electrical conductivity and the stimulation of cell differentiation of the stem cells. In addition, the chitosan scaffolds can significantly improve angiogenesis in the infarcted tissue by stimulating the production of the glycoprotein receptors of the vascular endothelial growth factor (VEGF) family. Therefore, the possible mechanisms of action of the chitosan scaffolds on cardiomyocytes and stem cells were analyzed. For all the advantages observed, it is considered that the treatment of MI with the chitosan scaffolds is promising, showing multiple advantages within the regenerative therapies of CVD.
Collapse
Affiliation(s)
- Bryan Beleño Acosta
- Grupo de Investigación de Fotoquímica y Fotobiología, Química, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| | - Rigoberto C. Advincula
- Department of Chemical and Biomolecular Engineering, University of Tennessee, Knoxville, TN 37996, USA
- Center for Nanophase Materials Sciences (CNMS), Oak Ridge National Laboratory, Oak Ridge, TN 37830, USA
| | - Carlos David Grande-Tovar
- Grupo de Investigación de Fotoquímica y Fotobiología, Química, Universidad del Atlántico, Carrera 30 Número 8-49, Puerto Colombia 081008, Colombia
| |
Collapse
|
164
|
Ma Z, Hua J, Liu J, Zhang B, Wang W, Yu X, Xu J. Mesenchymal Stromal Cell-Based Targeted Therapy Pancreatic Cancer: Progress and Challenges. Int J Mol Sci 2023; 24:ijms24043559. [PMID: 36834969 PMCID: PMC9966548 DOI: 10.3390/ijms24043559] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/18/2023] [Accepted: 02/01/2023] [Indexed: 02/12/2023] Open
Abstract
Pancreatic cancer is an aggressive malignancy with high mortality rates and poor prognoses. Despite rapid progress in the diagnosis and treatment of pancreatic cancer, the efficacy of current therapeutic strategies remains limited. Hence, better alternative therapeutic options for treating pancreatic cancer need to be urgently explored. Mesenchymal stromal cells (MSCs) have recently received much attention as a potential therapy for pancreatic cancer owing to their tumor-homing properties. However, the specific antitumor effect of MSCs is still controversial. To this end, we aimed to focus on the potential anti-cancer treatment prospects of the MSC-based approach and summarize current challenges in the clinical application of MSCs to treat pancreatic cancer.
Collapse
Affiliation(s)
- Zhilong Ma
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
- Correspondence: (X.Y.); (J.X.); Tel.: +86-021-64175590 (X.Y.); +86-021-64031446 (J.X.)
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong’An Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong’An Road, Shanghai 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
- Correspondence: (X.Y.); (J.X.); Tel.: +86-021-64175590 (X.Y.); +86-021-64031446 (J.X.)
| |
Collapse
|
165
|
Jerkic M, Szaszi K, Laffey JG, Rotstein O, Zhang H. Key Role of Mesenchymal Stromal Cell Interaction with Macrophages in Promoting Repair of Lung Injury. Int J Mol Sci 2023; 24:ijms24043376. [PMID: 36834784 PMCID: PMC9965074 DOI: 10.3390/ijms24043376] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/30/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Lung macrophages (Mφs) are essential for pulmonary innate immunity and host defense due to their dynamic polarization and phenotype shifts. Mesenchymal stromal cells (MSCs) have secretory, immunomodulatory, and tissue-reparative properties and have shown promise in acute and chronic inflammatory lung diseases and in COVID-19. Many beneficial effects of MSCs are mediated through their interaction with resident alveolar and pulmonary interstitial Mφs. Bidirectional MSC-Mφ communication is achieved through direct contact, soluble factor secretion/activation, and organelle transfer. The lung microenvironment facilitates MSC secretion of factors that result in Mφ polarization towards an immunosuppressive M2-like phenotype for the restoration of tissue homeostasis. M2-like Mφ in turn can affect the MSC immune regulatory function in MSC engraftment and tissue reparatory effects. This review article highlights the mechanisms of crosstalk between MSCs and Mφs and the potential role of their interaction in lung repair in inflammatory lung diseases.
Collapse
Affiliation(s)
- Mirjana Jerkic
- The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1T8, Canada
- Correspondence:
| | - Katalin Szaszi
- The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - John G. Laffey
- The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1T8, Canada
- Anaesthesia and Intensive Care Medicine, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Ori Rotstein
- The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Haibo Zhang
- The Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Unity Health Toronto, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Anesthesiology and Pain Medicine, Interdepartmental Division of Critical Care Medicine and Department of Physiology, University of Toronto, Toronto, ON M5G 1E2, Canada
| |
Collapse
|
166
|
Ríos S, Álvarez S, Smith PC, Sáez CG, Andrade C, Pinto N, Martínez CE. Smoking habits do not affect biological responses induced by leucocyte and platelet-rich fibrin in periodontal ligament cells. J Oral Pathol Med 2023; 52:169-180. [PMID: 36258298 DOI: 10.1111/jop.13374] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVE Leucocyte- and platelet-rich fibrin has been developed to stimulate wound healing response. However, it is currently unknown whether smoking affects the biological responses elicited by leucocyte- and platelet-rich fibrin on periodontal ligament-derived mesenchymal stromal cells. This study analyzes the kinetics of biomolecule release from leucocyte- and platelet-rich fibrin derived from smokers and nonsmokers and their effect on periodontal ligament cell proliferation and migration as essential biological activities during wound healing. METHODS Biomolecules present in leucocyte- and platelet-rich fibrin exudates and conditioned media collected from smokers and nonsmokers were analyzed by Luminex arrays. Periodontal ligament-derived mesenchymal stromal cell obtained from one nonsmoker were treated with leucocyte- and platelet-rich fibrin exudates or leucocyte- and platelet-rich fibrin conditioned media derived from both smokers and nonsmokers. The parameters evaluated included cell proliferation, determined by Ki67 immunostaining and migration assessed using transwell assays. Also, cells were treated with nicotine in the presence of fetal bovine serum 10% or leucocyte- and platelet-rich fibrin conditioned media. RESULTS A similar biomolecular profile was detected in leucocyte- and platelet-rich fibrin exudates and leucocyte- and platelet-rich fibrin conditioned media from smokers and nonsmokers, stimulating (periodontal ligament-derived mesenchymal stromal cell) proliferation, and migration to a comparable degree. Nicotine reduced cell proliferation and migration of periodontal cells; however, this effect was recovered in the presence of leucocyte- and platelet-rich fibrin conditioned media. CONCLUSION Leucocyte- and platelet-rich fibrin derived from smokers could be an autologous source of biomolecules to stimulate cell biological activities involved in wound healing in smokers who have difficulties in ceasing this habit. Clinical trials are required to evaluate the impact of leucocyte- and platelet-rich fibrin on healing responses in smokers.
Collapse
Affiliation(s)
- Susana Ríos
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Simón Álvarez
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricio C Smith
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia G Sáez
- School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Nelson Pinto
- Faculty of Dentistry, Universidad de los Andes, Santiago, Chile
| | - Constanza E Martínez
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
167
|
Li SD, Xing W, Wang SC, Li YB, Jiang H, Zheng HX, Li XM, Yang J, Guo DB, Xie XY, Jiang RQ, Fan C, Li L, Xu X, Fei J. Fibulin2: a negative regulator of BMSC osteogenic differentiation in infected bone fracture healing. Exp Mol Med 2023; 55:443-456. [PMID: 36797542 PMCID: PMC9981700 DOI: 10.1038/s12276-023-00942-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/26/2022] [Accepted: 12/02/2022] [Indexed: 02/18/2023] Open
Abstract
Bone fracture remains a common occurrence, with a population-weighted incidence of approximately 3.21 per 1000. In addition, approximately 2% to 50% of patients with skeletal fractures will develop an infection, one of the causes of disordered bone healing. Dysfunction of bone marrow mesenchymal stem cells (BMSCs) plays a key role in disordered bone repair. However, the specific mechanisms underlying BMSC dysfunction caused by bone infection are largely unknown. In this study, we discovered that Fibulin2 expression was upregulated in infected bone tissues and that BMSCs were the source of infection-induced Fibulin2. Importantly, Fibulin2 knockout accelerated mineralized bone formation during skeletal development and inhibited inflammatory bone resorption. We demonstrated that Fibulin2 suppressed BMSC osteogenic differentiation by binding to Notch2 and inactivating the Notch2 signaling pathway. Moreover, Fibulin2 knockdown restored Notch2 pathway activation and promoted BMSC osteogenesis; these outcomes were abolished by DAPT, a Notch inhibitor. Furthermore, transplanted Fibulin2 knockdown BMSCs displayed better bone repair potential in vivo. Altogether, Fibulin2 is a negative regulator of BMSC osteogenic differentiation that inhibits osteogenesis by inactivating the Notch2 signaling pathway in infected bone.
Collapse
Affiliation(s)
- Shi-Dan Li
- Department of Orthopaedics, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Wei Xing
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Shao-Chuan Wang
- Department of Orthopaedics, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - You-Bin Li
- Department of Orthopaedics, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Hao Jiang
- Department of Orthopaedics, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Han-Xuan Zheng
- Department of Nursing, Montreal Neurological Hospital, 3801 Rue University, Montréal, QC H3A 2B4, Canada
| | - Xiao-Ming Li
- Department of Military Traffic Injury Prevention, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Jing Yang
- Department of Emergency, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - De-Bin Guo
- Department of Emergency, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Xiao-Yu Xie
- Department of Orthopaedics, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Ren-Qing Jiang
- Department for Combat Casualty Care Training, Training Base for Army Health Care, Third Military Medical University, Chongqing, 400042, People's Republic of China
| | - Chao Fan
- Medical Research Center, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Lei Li
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Xiang Xu
- Department of Stem Cell and Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| | - Jun Fei
- Department of Emergency, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
168
|
Qin L, Liu N, Bao CLM, Yang DZ, Ma GX, Yi WH, Xiao GZ, Cao HL. Mesenchymal stem cells in fibrotic diseases-the two sides of the same coin. Acta Pharmacol Sin 2023; 44:268-287. [PMID: 35896695 PMCID: PMC9326421 DOI: 10.1038/s41401-022-00952-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Fibrosis is caused by extensive deposition of extracellular matrix (ECM) components, which play a crucial role in injury repair. Fibrosis attributes to ~45% of all deaths worldwide. The molecular pathology of different fibrotic diseases varies, and a number of bioactive factors are involved in the pathogenic process. Mesenchymal stem cells (MSCs) are a type of multipotent stem cells that have promising therapeutic effects in the treatment of different diseases. Current updates of fibrotic pathogenesis reveal that residential MSCs may differentiate into myofibroblasts which lead to the fibrosis development. However, preclinical and clinical trials with autologous or allogeneic MSCs infusion demonstrate that MSCs can relieve the fibrotic diseases by modulating inflammation, regenerating damaged tissues, remodeling the ECMs, and modulating the death of stressed cells after implantation. A variety of animal models were developed to study the mechanisms behind different fibrotic tissues and test the preclinical efficacy of MSC therapy in these diseases. Furthermore, MSCs have been used for treating liver cirrhosis and pulmonary fibrosis patients in several clinical trials, leading to satisfactory clinical efficacy without severe adverse events. This review discusses the two opposite roles of residential MSCs and external MSCs in fibrotic diseases, and summarizes the current perspective of therapeutic mechanism of MSCs in fibrosis, through both laboratory study and clinical trials.
Collapse
Affiliation(s)
- Lei Qin
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Nian Liu
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Chao-le-meng Bao
- CASTD Regengeek (Shenzhen) Medical Technology Co. Ltd, Shenzhen, 518000 China
| | - Da-zhi Yang
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Gui-xing Ma
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Wei-hong Yi
- grid.33199.310000 0004 0368 7223Department of Orthopedics, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518000 China
| | - Guo-zhi Xiao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| | - Hui-ling Cao
- grid.263817.90000 0004 1773 1790Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, 518055 China
| |
Collapse
|
169
|
Infusion of Some but Not All Types of Human Perinatal Stromal Cells Prevent Organ Fibrosis in a Humanized Graft versus Host Disease Murine Model. Biomedicines 2023; 11:biomedicines11020415. [PMID: 36830951 PMCID: PMC9953740 DOI: 10.3390/biomedicines11020415] [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: 01/12/2023] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
Allogeneic transplant rejection represents a medical complication that leads to high morbidity and mortality rates. There are no treatments to effectively prevent fibrosis; however, there is great interest in evaluating the use of perinatal mesenchymal stromal cells (MSCs) and other MSCs to prevent fibrosis associated with chronic rejection. In this study, we isolated human perinatal stromal cells (PSCs) from amnion (AM-PSC), placental villi (PV-PSC), and umbilical cord (UC-PSC) tissues, demonstrating the phenotypic characteristics of MSCs as well as a >70% expression of the immunomodulatory markers CD273 and CD210. The administration of a single dose (250,000 cells) of each type of PSC in a humanized graft versus host disease (hGvHD) NSG® murine model delayed the progression of the disease as displayed by weight loss and GvHD scores ranging at various levels without affecting the hCD3+ population. However, only PV-PSCs demonstrated an increased survival rate of 50% at the end of the study. Furthermore, a histopathological evaluation showed that only PV-PSC cells could reduce human CD45+ cell infiltration and the fibrosis of the lungs and liver. These findings indicate that not all PSCs have similar therapeutic potential, and that PV-PSC as a cell therapeutic may have an advantage for targeting fibrosis related to allograft rejection.
Collapse
|
170
|
Behm C, Blufstein A, Gahn J, Moritz A, Rausch-Fan X, Andrukhov O. 25-hydroxyvitamin D 3 generates immunomodulatory plasticity in human periodontal ligament-derived mesenchymal stromal cells that is inflammatory context-dependent. Front Immunol 2023; 14:1100041. [PMID: 36761739 PMCID: PMC9902380 DOI: 10.3389/fimmu.2023.1100041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Introduction Human periodontal ligament-derived mesenchymal stromal cells (hPDL-MSCs) exhibit a tight bi-directional interaction with CD4+ T lymphocytes. The hPDL-MSCs' immunomodulatory abilities are drastically enhanced by pro-inflammatory cytokines via boosting the expression of various immunomediators. 25-hydroxyvitamin D3 (25(OH)D3), the major metabolite of vitamin D3 in the blood, affects both hPDL-MSCs and CD4+ T lymphocytes, but its influence on their interaction is unknown. Methods Therefore, primary hPDL-MSCs were stimulated in vitro with tumor necrosis factor (TNF)-α a or interleukin (IL)-1β in the absence and presence of 25(OH)D3 followed by an indirect co-culture with phytohemagglutinin-activated CD4+ T lymphocytes. The CD4+ T lymphocyte proliferation, viability, and cytokine secretion were analyzed. Additionally, the expression of various immunomediators in hPDL-MSCs was investigated, and their implication was verified by using pharmacological inhibitors. Results 25(OH)D3 significantly counteracted the suppressive effects of IL-1β-treated hPDL-MSCs on CD4+ T lymphocyte proliferation, whereas no effects were observed in the presence of TNF-α. Additionally, 25(OH)D3 significantly increased the percentage of viable CD4+ T lymphocytes via TNF-α- or IL-1β-treated hPDL-MSCs. It also caused a significant decrease in interferon-γ, IL-17A, and transforming growth factor-β productions, which were triggered by TNF-α-treated hPDL-MSCs. 25(OH)D3 significantly decreased the production of various immunomediators in hPDL-MSCs. Inhibition of two of them, prostaglandin E2 and indoleamine-2,3-dioxygenase-1, partially abolished some of the hPDL-MSCs-mediated effects of 25(OH)D3 on CD4+ T lymphocytes. Conclusion These data indicate that 25(OH)D3 influences the immunomodulatory activities of hPDL-MSCs. This modulatory potential seems to have high plasticity depending on the local cytokine conditions and may be involved in regulating periodontal tissue inflammatory processes.
Collapse
Affiliation(s)
- Christian Behm
- Competence Center Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Alice Blufstein
- Competence Center Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Clinical Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Johannes Gahn
- Competence Center Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Andreas Moritz
- Clinical Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Xiaohui Rausch-Fan
- Clinical Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Center for Clinical Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Oleh Andrukhov
- Competence Center Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
171
|
Xue J, Liu Y. Mesenchymal Stromal/Stem Cell (MSC)-Based Vector Biomaterials for Clinical Tissue Engineering and Inflammation Research: A Narrative Mini Review. J Inflamm Res 2023; 16:257-267. [PMID: 36713049 PMCID: PMC9875582 DOI: 10.2147/jir.s396064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) have the ability of self-renewal, the potential of multipotent differentiation, and a strong paracrine capacity, which are mainly used in the field of clinical medicine including dentistry and orthopedics. Therefore, tissue engineering research using MSCs as seed cells is a current trending directions. However, the healing effect of direct cell transplantation is unstable, and the paracrine/autocrine effects of MSCs cannot be effectively elicited. Tumorigenicity and heterogeneity are also concerns. The combination of MSCs as seed cells and appropriate vector materials can form a stable cell growth environment, maximize the secretory features of stem cells, and improve the biocompatibility and mechanical properties of vector materials that facilitate the delivery of drugs and various secretory factors. There are numerous studies on tissue engineering and inflammation of various biomaterials, mainly involving bioceramics, alginate, chitosan, hydrogels, cell sheets, nanoparticles, and three-dimensional printing. The combination of bioceramics, hydrogels and cell sheets with stem cells has demonstrated good therapeutic effects in clinical applications. The application of alginate, chitosan, and nanoparticles in animal models has also shown good prospects for clinical applications. Three-dimensional printing technology can circumvent the shortage of biomaterials, greatly improve the properties of vector materials, and facilitate the transplantation of MSCs. The purpose of this narrative review is to briefly discuss the current use of MSC-based carrier biomaterials to provide a useful resource for future tissue engineering and inflammation research using stem cells as seed cells.
Collapse
Affiliation(s)
- Junshuai Xue
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Yang Liu
- Department of General Surgery, Vascular Surgery, Qilu Hospital of Shandong University, Jinan City, People’s Republic of China,Correspondence: Yang Liu, Department of General surgery, Vascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, People’s Republic of China, Tel +86 18560088317, Email
| |
Collapse
|
172
|
Cicione C, Vadalà G, Di Giacomo G, Tilotta V, Ambrosio L, Russo F, Zampogna B, Cannata F, Papalia R, Denaro V. Micro-fragmented and nanofat adipose tissue derivatives: In vitro qualitative and quantitative analysis. Front Bioeng Biotechnol 2023; 11:911600. [PMID: 36733959 PMCID: PMC9887143 DOI: 10.3389/fbioe.2023.911600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Introduction: Adipose tissue is widely exploited in regenerative medicine thanks to its trophic properties, mainly based on the presence of adipose-derived stromal cells. Numerous devices have been developed to promote its clinical use, leading to the introduction of one-step surgical procedures to obtain minimally manipulated adipose tissue derivatives. However, only a few studies compared their biological properties. This study aimed to characterize micro-fragmented (MAT) and nanofat adipose tissue (NAT) obtained with two different techniques. Methods: MAT, NAT and unprocessed lipoaspirate were collected from surgical specimens. RNA extraction and collagenase isolation of stromal vascular fraction (SVF) were performed. Tissue sections were analysed by histological and immunohistochemical (collagen type I, CD31, CD34 and PCNA) staining to assess tissue morphology and cell content. qPCR was performed to evaluate the expression of stemness-related (SOX2, NANOG and OCT3/4), extracellular matrix (COL1A1) and inflammatory genes (IL1β, IL6 and iNOS). Furthermore, multilineage differentiation was assessed following culture in adipogenic and osteogenic media and staining with Oil Red O and Alizarin red. ASC immunophenotype was assessed by flow cytometric analysis of CD90, CD105, CD73 and CD45. Results: Histological and immunohistochemical results showed an increased amount of stroma and a reduction of adipocytes in MAT and NAT, with the latter displaying the highest content of collagen type I, CD31, CD34 and PCNA. From LA to MAT and NAT, an increasing expression of NANOG, SOX2, OCT3/4, COL1A1 and IL6 was noted, while no significant differences in terms of IL1β and iNOS emerged. No statistically significant differences were noted between NAT and SVF in terms of stemness-related genes, while the latter demonstrated a significantly higher expression of stress-related markers. SVF cells derived from all three samples (LA, MAT, and NAT) showed a similar ASC immunoprofile as well as osteogenic and adipogenic differentiation. Discussion: Our results showed that both MAT and NAT techniques allowed the rapid isolation of ASC-rich grafts with a high anabolic and proliferative potential. However, NAT showed the highest levels of extracellular matrix content, replicating cells, and stemness gene expression. These results may provide precious clues for the use of adipose tissue derivatives in the clinical setting.
Collapse
Affiliation(s)
- Claudia Cicione
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gianluca Vadalà
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy,*Correspondence: Gianluca Vadalà,
| | - Giuseppina Di Giacomo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Veronica Tilotta
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Luca Ambrosio
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Fabrizio Russo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Biagio Zampogna
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesca Cannata
- Operative Research Unit of Endocrinology and Diabetes, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rocco Papalia
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy,Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Vincenzo Denaro
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
173
|
Cheng HY, Anggelia MR, Lin CH, Wei FC. Toward transplantation tolerance with adipose tissue-derived therapeutics. Front Immunol 2023; 14:1111813. [PMID: 37187733 PMCID: PMC10175575 DOI: 10.3389/fimmu.2023.1111813] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Solid organ and composite tissue allotransplanation have been widely applied to treat end-stage organ failure and massive tissue defects, respectively. Currently there are a lot of research endeavors focusing on induction of transplantation tolerance, to relieve the burden derived from long-term immunosuppressant uptake. The mesenchymal stromal cells (MSCs) have been demonstrated with potent immunomodulatory capacities and applied as promising cellular therapeutics to promote allograft survival and induce tolerance. As a rich source of adult MSCs, adipose tissue provides additional advantages of easy accessibility and good safety profile. In recent years, the stromal vascular fraction (SVF) isolated from adipose tissues following enzymatic or mechanical processing without in vitro culture and expansion has demonstrated immunomodulatory and proangiogenic properties. Furthermore, the secretome of AD-MSCs has been utilized in transplantation field as a potential "cell-free" therapeutics. This article reviews recent studies that employ these adipose-derived therapeutics, including AD-MSCs, SVF, and secretome, in various aspects of organ and tissue allotransplantation. Most reports validate their efficacies in prolonging allograft survival. Specifically, the SVF and secretome have performed well for graft preservation and pretreatment, potentially through their proangiogenic and antioxidative capacities. In contrast, AD-MSCs were suitable for peri-transplantation immunosuppression. The proper combination of AD-MSCs, lymphodepletion and conventional immunosuppressants could consistently induce donor-specific tolerance to vascularized composite allotransplants (VCA). For each type of transplantation, optimizing the choice of therapeutics, timing, dose, and frequency of administration may be required. Future progress in the application of adipose-derived therapeutics to induce transplantation tolerance will be further benefited by continued research into their mechanisms of action and the development of standardized protocols for isolation methodologies, cell culture, and efficacy evaluation.
Collapse
Affiliation(s)
- Hui-Yun Cheng
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- *Correspondence: Hui-Yun Cheng,
| | - Madonna Rica Anggelia
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Cheng-Hung Lin
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chan Wei
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
174
|
Huang Z, Su X, Julaiti M, Chen X, Luan Q. The role of PRX1-expressing cells in periodontal regeneration and wound healing. Front Physiol 2023; 14:978640. [PMID: 36960156 PMCID: PMC10027693 DOI: 10.3389/fphys.2023.978640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
The ideal outcome of wound healing is the complete restoration of the structure and function of the original tissue. Stem cells are one of the key factors in this process. Currently, the strategy of periodontal regeneration based on mesenchymal stem cells (MSCs) is generally used to expand stem cells in vitro and then transplant them in vivo. However, their clinical application is limited. In fact, the human body has the capacity to regenerate through stem cells residing in different tissues, even without external therapeutic intervention. Stem cell niches are present in many adult tissues, such as the periodontal ligament and gingiva, and stem cells might remain in a quiescent state in their niches until they are activated in response to a regenerative need. Activated stem cells can exit the niche and proliferate, self-renew, and differentiate to regenerate original structures. Thus, harnessing the regenerative potential of endogenous stem cells in situ has gained increasing attention as a simpler, safer, and more applicable alternative to stem cell transplantation. Nevertheless, there are several key problems to be solved in the application of periodontal mesenchymal stem cells. Thus, animal studies will be especially important to deepen our knowledge of the in vivo mechanisms of mesenchymal stem cells. Studies with conditional knockout mice, in which the expression of different proteins can be eliminated in a tissue-specific manner, are especially important. Post-natal cells expressing the paired-related homeobox protein 1 (PRX1 or PRRX1), a transcription factor expressed in the mesenchyme during craniofacial and limb development, have been shown to have characteristics of skeletal stem cells. Additionally, following wounding, dermal Prx1+ cells are found out of their dermal niches and contribute to subcutaneous tissue repair. Postnatal Prx1+ cells are uniquely injury-responsive. Meanwhile, current evidence shows that Prx1+ cells contribute to promote dentin formation, wound healing of alveolar bone and formation of mouse molar and periodontal ligament. Initial result of our research group also indicates Prx1-expressing cells in bone tissue around the punch wound area of gingiva increased gradually. Collectively, this review supports the future use of PRX1 cells to stimulate their potential to play an important role in endogenous regeneration during periodontal therapy.
Collapse
Affiliation(s)
- Zhen Huang
- Beijing Key Laboratory of Digital Stomatology, NMPA Key Laboratory for Dental Materials, Department of Periodontology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xu Su
- Department of Stomatology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Miliya Julaiti
- Department of Stomatology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Xiaotao Chen
- Department of Stomatology, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
- *Correspondence: Xiaotao Chen, ; Qingxian Luan,
| | - Qingxian Luan
- Beijing Key Laboratory of Digital Stomatology, NMPA Key Laboratory for Dental Materials, Department of Periodontology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, Peking University School and Hospital of Stomatology, Beijing, China
- *Correspondence: Xiaotao Chen, ; Qingxian Luan,
| |
Collapse
|
175
|
Zhang Y, Xu X, Cheng H, Zhou F. AIM2 and Psoriasis. Front Immunol 2023; 14:1085448. [PMID: 36742336 PMCID: PMC9889639 DOI: 10.3389/fimmu.2023.1085448] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Psoriasis is a chronic inflammatory skin disease occurring worldwide, with multiple systemic complications, which seriously affect the quality of life and physical and mental health of patients. The pathogenesis of psoriasis is related to the environment, genetics, epigenetics, and dysregulation of immune cells such as T cells, dendritic cells (DCs), and nonimmune cells such as keratinocytes. Absent in melanoma 2 (AIM2), a susceptibility gene locus for psoriasis, has been strongly linked to the genetic and epigenetic aspects of psoriasis and increased in expression in psoriatic keratinocytes. AIM2 was found to be activated in an inflammasome-dependent way to release IL-1β and IL-18 to mediate inflammation, and to participate in immune regulation in psoriasis, or in an inflammasome-independent way by regulating the function of regulatory T(Treg) cells or programming cell death in keratinocytes as well as controlling the proliferative state of different cells. AIM2 may also play a role in the recurrence of psoriasis by trained immunity. In this review, we will elaborate on the characteristics of AIM2 and how AIM2 mediates the development of psoriasis.
Collapse
Affiliation(s)
- Yuxi Zhang
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Xiaoqing Xu
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Hui Cheng
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| | - Fusheng Zhou
- Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China.,Institute of Dermatology, Anhui Medical University, Hefei, Anhui, China.,Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China
| |
Collapse
|
176
|
Torrents S, Grau-Vorster M, Vives J. Potency Assays: The 'Bugaboo' of Stem Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:29-38. [PMID: 37258782 DOI: 10.1007/978-3-031-30040-0_3] [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
Substantially manipulated cell-based products for human use are considered medicines and therefore regulatory authorities require extensive characterisation in terms of identity, purity and potency. The latter critical quality attribute is probably the most challenging to identify and measure, requiring provision that potency assays should reflect the intended mechanism of action and demonstrate the drugs' biological effect. However, in most cases, the mechanisms involved are not fully understood, making the definition and validation of suitable potency tests difficult, a 'bugaboo' quest to be feared. Although it is evident that much work is still needed in the scientific arena, the present chapter focuses on strategies currently used by developers of cell- and gene-based therapies to demonstrate potency of innovative medicines, the regulatory framework and need for standardisation seeking to demystify critical factors to consider when designing a potency assay.
Collapse
Affiliation(s)
- Sílvia Torrents
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marta Grau-Vorster
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain
- Transfusion Medicine group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joaquim Vives
- Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain.
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
177
|
Chinnadurai R, Viswanathan S, Moll G. Editorial: Next generation MSC therapy manufacturing, potency and mechanism of action analysis. Front Immunol 2023; 14:1192636. [PMID: 37153609 PMCID: PMC10161792 DOI: 10.3389/fimmu.2023.1192636] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, United States
- *Correspondence: Raghavan Chinnadurai, ; Sowmya Viswanathan, ; Guido Moll,
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, Division of Hematology, Department of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: Raghavan Chinnadurai, ; Sowmya Viswanathan, ; Guido Moll,
| | - Guido Moll
- BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies (BSRT), Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, all Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health (BIH), Berlin, Germany
- *Correspondence: Raghavan Chinnadurai, ; Sowmya Viswanathan, ; Guido Moll,
| |
Collapse
|
178
|
Hackel A, Vollmer S, Bruderek K, Lang S, Brandau S. Immunological priming of mesenchymal stromal/stem cells and their extracellular vesicles augments their therapeutic benefits in experimental graft-versus-host disease via engagement of PD-1 ligands. Front Immunol 2023; 14:1078551. [PMID: 36875112 PMCID: PMC9978482 DOI: 10.3389/fimmu.2023.1078551] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/09/2023] [Indexed: 02/18/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) and their extracellular vesicles (EVs) exert profound anti-inflammatory and regenerative effects in inflammation and tissue damage, which makes them an attractive tool for cellular therapies. In this study we have assessed the inducible immunoregulatory properties of MSCs and their EVs upon stimulation with different combinations of cytokines. First, we found that MSCs primed with IFN-γ, TNF-α and IL-1β, upregulate the expression of PD-1 ligands, as crucial mediators of their immunomodulatory activity. Further, primed MSCs and MSC-EVs, compared to unstimulated MSCs and MSC-EVs, had increased immunosuppressive effects on activated T cells and mediated an enhanced induction of regulatory T cells, in a PD-1 dependent manner. Importantly, EVs derived from primed MSCs reduced the clinical score and prolonged the survival of mice in a model of graft-versus-host disease. These effects could be reversed in vitro and in vivo by adding neutralizing antibodies directed against PD-L1 and PD-L2 to both, MSCs and their EVs. In conclusion, our data reveal a priming strategy that potentiates the immunoregulatory function of MSCs and their EVs. This concept also provides new opportunities to improve the clinical applicability and efficiency of cellular or EV-based therapeutic MSC products.
Collapse
Affiliation(s)
- Alexander Hackel
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Vollmer
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kirsten Bruderek
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Lang
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sven Brandau
- Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
179
|
Marson RF, Regner AP, da Silva Meirelles L. Mesenchymal "stem" cells, or facilitators for the development of regenerative macrophages? Pericytes at the interface of wound healing. Front Cell Dev Biol 2023; 11:1148121. [PMID: 36936686 PMCID: PMC10017474 DOI: 10.3389/fcell.2023.1148121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Cultured mesenchymal stromal cells are among the most used cells in clinical trials. Currently, their potential benefits include provision of mature cell types through differentiation, and secretion of various types of paracrine signaling molecules. Even though research on these cells has spanned some decades now, surprisingly, their therapeutic potential has not been fully translated into clinical practice yet, which calls for further understanding of their intrinsic nature and modes of action. In this review, after discussing pieces of evidence that suggest that some perivascular cells may exhibit mesenchymal stem cell characteristics in vivo, we examine the possibility that subpopulations of perivascular and/or adventitial cells activated after tissue injury behave as MSCs and contribute to the resolution of tissue injury by providing cues for the development of regenerative macrophages at injured sites. Under this perspective, an important contribution of cultured MSCs (or their acellular products, such as extracellular vesicles) used in cell therapies would be to instigate the development of M2-like macrophages that support the tissue repair process.
Collapse
Affiliation(s)
- Renan Fava Marson
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
| | - Andrea Pereira Regner
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
| | - Lindolfo da Silva Meirelles
- Graduate Program in Cellular and Molecular Biology Applied to Health—PPGBioSaúde, Lutheran University of Brazil, Canoas, Brazil
- School of Medicine, Lutheran University of Brazil, Canoas, Brazil
- *Correspondence: Lindolfo da Silva Meirelles, ,
| |
Collapse
|
180
|
Miclau K, Hambright WS, Huard J, Stoddart MJ, Bahney CS. Cellular expansion of MSCs: Shifting the regenerative potential. Aging Cell 2023; 22:e13759. [PMID: 36536521 PMCID: PMC9835588 DOI: 10.1111/acel.13759] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/14/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal-derived stromal or progenitor cells, commonly called "MSCs," have attracted significant clinical interest for their remarkable abilities to promote tissue regeneration and reduce inflammation. Recent studies have shown that MSCs' therapeutic effects, originally attributed to the cells' direct differentiation capacity into the tissue of interest, are largely driven by the biomolecules the cells secrete, including cytokines, chemokines, growth factors, and extracellular vesicles containing miRNA. This secretome coordinates upregulation of endogenous repair and immunomodulation in the local microenvironment through crosstalk of MSCs with host tissue cells. Therapeutic applications for MSCs and their secretome-derived products often involve in vitro monolayer expansion. However, consecutive passaging of MSCs significantly alters their therapeutic potential, inducing a broad shift from a pro-regenerative to a pro-inflammatory phenotype. A consistent by-product of in vitro expansion of MSCs is the onset of replicative senescence, a state of cell arrest characterized by an increased release of proinflammatory cytokines and growth factors. However, little is known about changes in the secretome profile at different stages of in vitro expansion. Some culture conditions and bioprocessing techniques have shown promise in more effectively retaining the pro-regenerative and anti-inflammatory MSC phenotype throughout expansion. Understanding how in vitro expansion conditions influence the nature and function of MSCs, and their associated secretome, may provide key insights into the underlying mechanisms driving these alterations. Elucidating the dynamic and diverse changes in the MSC secretome at each stage of in vitro expansion is a critical next step in the development of standardized, safe, and effective MSC-based therapies.
Collapse
Affiliation(s)
- Katherine Miclau
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
- Orthopaedic Trauma Institute (OTI)University of California San FranciscoSan FranciscoCaliforniaUSA
| | - William S. Hambright
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
| | - Martin J. Stoddart
- Orthopaedic Trauma Institute (OTI)University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Chelsea S. Bahney
- Center for Regenerative and Personalized Medicine (CRPM)Steadman Philippon Research InstituteVailColoradoUSA
- AO Research Institute DavosDavosSwitzerland
| |
Collapse
|
181
|
Burns JS. The Art of Stem Cell-Based Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:1-12. [PMID: 37258780 DOI: 10.1007/978-3-031-30040-0_1] [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
Potency assays represent crucial experiments at the hub of the comprehensive complexity surrounding cell therapy. Moreover, numerous factors beyond biological and scientific considerations are involved in achieving successful potency assays that fulfil regulatory authority approval for a new advanced therapy medicinal product. Though this can mean a frustratingly long period of discovery and development, progress in cell therapy is nowadays proceeding remarkably quickly, assisted by the potency assay rigorously placing emphasis on the need to critically analyse the key factor/s responsible for the therapeutic mechanism of action. History has shown that it can take many decades for there to be an improved understanding of a mechanism of action. Yet the chasing of precise targets has revolutionised medicine, with no clearer example than approaches to viral pandemics. The centuries involved in the eradication of smallpox have paved the way for an unprecedented pace of vaccine development for the Covid-19 pandemic. Such extraordinary accomplishments foster encouragement that similarly for stem cell-based therapy, our scientific knowledge will continue to improve apace. This chapter focuses on the art of experimentation and discovery, introducing potency assay requisites and numerous factors that can influence potency assay outcomes. A comprehensive understanding of potency assays and their development can hasten the provision of new cell therapies to help resolve burdensome diseases of unmet medical need.
Collapse
Affiliation(s)
- Jorge S Burns
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
182
|
Soltero-Rivera M, Hart S, Blandino A, Vapniarsky N, Arzi B. Mesenchymal stromal cell therapy for feline chronic gingivostomatitis: Long term experience. Front Vet Sci 2023; 10:1171922. [PMID: 37168097 PMCID: PMC10165997 DOI: 10.3389/fvets.2023.1171922] [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: 02/22/2023] [Accepted: 03/23/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction Mesenchymal stromal cells (MSC) therapy has emerged as a potential treatment option for refractory FCGS. However, there is a lack of long-term data on the use of MSC therapy in cats. This study aimed to evaluate the long-term safety and efficacy of MSC therapy for FCGS and investigate potential factors associated with treatment outcomes. Methods This study was a retrospective evaluation of 38 client-owned cats with refractory FCGS who received MSC therapy. Medical records, histopathology, and the Stomatitis Activity Disease Index (SDAI) were reviewed. Correlations of the long-term follow-up success rates with SDAI and cell line type used were conducted. A client survey was also performed to assess side effect occurrence, quality-of-life following treatment, and overall treatment satisfaction. Results Long-term follow-up ranged from 2 to 9 years post-MSC treatment. The overall positive response rate to MSC treatment was 65.5%, with 58.6% of cats exhibiting permanent improvement or cure. Adverse effects occurring during or immediately after treatment were noted in 34.2% of cases, the majority being transient, self-resolving transfusion-like reactions. No long-term adverse events were noted. No significant correlation in outcome was detected between allogeneic and autologous MSC treatment (p = 0.871) or the severity of the SDAI at entry (p = 0.848) or exit (p = 0.166), or the delta SDAI between entry and exit (p = 0.178). The status 6 months (none to partial improvement vs. substantial improvement to resolution) post-therapy was a predictor of long-term response (value of p < 0.041). Most clients were satisfied with the treatment and outcomes, with 90.6% willing to pursue treatment again, given a similar situation. Discussion The results of this study support the use of both autologous and allogeneic MSC as an efficacious and safe therapeutic option for refractory FCGS.
Collapse
Affiliation(s)
- Maria Soltero-Rivera
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- *Correspondence: Maria Soltero-Rivera,
| | - Sterling Hart
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Andrew Blandino
- Department of Statistics, University of California, Davis, Davis, CA, United States
| | - Natalia Vapniarsky
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
183
|
Barreiro BOB, Koth VS, Sesterheim P, Salum FG, Rübensam G, Augustin AH, Cherubini K. Autogenous dentin combined with mesenchymal stromal cells as an alternative alveolar bone graft: an in vivo study. Clin Oral Investig 2022; 27:1907-1922. [PMID: 36574044 DOI: 10.1007/s00784-022-04840-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Considering the chemical and structural properties of dentin, this study was aimed at evaluating the effect of dentin matrix alone or combined with mesenchymal stromal cells (MSC) on postextraction alveolar bone regeneration. MATERIAL AND METHODS Wistar rats were subjected to tooth extraction with osteotomy and allocated into groups according to the graft inserted: (1) Gelita-Spon®, (2) Bio-Oss®, (3) Dentin, (4) MSC, (5) Dentin/MSC, and (6) Control. Maxillae were analyzed by means of hematoxylin and eosin (H&E) staining, immunohistochemical (IHC) analysis, microcomputed tomography (micro-CT), and scanning electron microscopy (SEM). Serum levels of calcium and phosphorus were quantified. RESULTS The Bio-Oss group showed less bone than Gelita-Spon and Dentin/MSC; no other significant differences were seen in H&E analysis. The Bio-Oss group showed higher expression of collagen type I compared to the Dentin and Dentin/MSC groups and also higher osteocalcin expression than the Dentin/MSC group. There was a tendency of higher expression of osteopontin in the MSC, Dentin, and Dentin/MSC groups and higher VEGF in the MSC group. On micro-CT analysis, the Bio-Oss and the Dentin/MSC groups exhibited greater bone volume than the Control. Serum calcium and phosphorus levels did not significantly differ between the groups. SEM analysis depicted particles of Bio-Oss and dentin in the respective groups, as well as significant cellularity in the MSC group. CONCLUSION Autogenous nondemineralized dentin is an alternative for alveolar bone grafting, which can be improved by combination with MSC. CLINICAL RELEVANCE This work provides support for the clinical applicability of dentin graft alone or combined with MSC.
Collapse
Affiliation(s)
- Bernardo Ottoni Braga Barreiro
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Valesca Sander Koth
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Patrícia Sesterheim
- Experimental Cardiology Center, Institute of Cardiology of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Gonçalves Salum
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Gabriel Rübensam
- Toxicology and Pharmacology Research Center (INTOX), School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Adolpho Herbert Augustin
- Institute of Petroleum and Natural Resources (IPR), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Karen Cherubini
- School of Health and Life Sciences, Post-Graduate Program in Dentistry, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Serviço de Estomatologia, Hospital São Lucas PUCRS, Av. Ipiranga, 6690 Sala 231, Bairro Jardim Botânico, CEP: 90.610-000, Porto Alegre, RS, Brazil.
| |
Collapse
|
184
|
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: 3] [Impact Index Per Article: 1.5] [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
|
185
|
Carbonised Human Hair Incorporated in Agar/KGM Bioscaffold for Tissue Engineering Application: Fabrication and Characterisation. Polymers (Basel) 2022; 14:polym14245489. [PMID: 36559856 PMCID: PMC9785055 DOI: 10.3390/polym14245489] [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: 11/13/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022] Open
Abstract
Carbon derived from biomass waste usage is rising in various fields of application due to its availability, cost-effectiveness, and sustainability, but it remains limited in tissue engineering applications. Carbon derived from human hair waste was selected to fabricate a carbon-based bioscaffold (CHAK) due to its ease of collection and inexpensive synthesis procedure. The CHAK was fabricated via gelation, rapid freezing, and ethanol immersion and characterised based on their morphology, porosity, Fourier transforms infrared (FTIR), tensile strength, swelling ability, degradability, electrical conductivity, and biocompatibility using Wharton’s jelly-derived mesenchymal stem cells (WJMSCs). The addition of carbon reduced the porosity of the bioscaffold. Via FTIR analysis, the combination of carbon, agar, and KGM was compatible. Among the CHAK, the 3HC bioscaffold displayed the highest tensile strength (62.35 ± 29.12 kPa). The CHAK also showed excellent swelling and water uptake capability. All bioscaffolds demonstrated a slow degradability rate (<50%) after 28 days of incubation, while the electrical conductivity analysis showed that the 3AHC bioscaffold had the highest conductivity compared to other CHAK bioscaffolds. Our findings also showed that the CHAK bioscaffolds were biocompatible with WJMSCs. These findings showed that the CHAK bioscaffolds have potential as bioscaffolds for tissue engineering applications.
Collapse
|
186
|
Transcription Factors STAT3 and MYC Are Key Players of Human Platelet Lysate-Induced Cell Proliferation. Int J Mol Sci 2022; 23:ijms232415782. [PMID: 36555426 PMCID: PMC9781157 DOI: 10.3390/ijms232415782] [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: 11/09/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022] Open
Abstract
Human platelet lysate (HPL) is an efficient alternative for animal serum supplements, significantly enhancing stromal cell proliferation. However, the molecular mechanism behind this growth-promoting effect remains elusive. The aim of this study was to investigate the effect of HPL on cell cycle gene expression in different human stromal cells and to identify the main key players that mediate HPL's growth-enhancing effect. RT-qPCR and an antibody array revealed significant upregulation of cell cycle genes in stromal cells cultured in HPL. As HPL is rich in growth factors that are ligands of tyrosine kinase receptor (TKR) pathways, we used TKR inhibitors and could significantly reduce cell proliferation. Genome profiling, RT-qPCR and Western blotting revealed an enhanced expression of the transcription factors signal transducer and activator of transcription 3 (STAT3) and MYC, both known TKR downstream effectors and stimulators of cell proliferation, in response to HPL. In addition, specifically blocking STAT3 resulted in reduced cell proliferation and expression of cell cycle genes. Our data indicate that HPL-enhanced cell proliferation can, at least in part, be explained by the TKR-enhanced expression of STAT3 and MYC, which in turn induce the expression of genes being involved in the promotion and control of the cell cycle.
Collapse
|
187
|
An efficient human stem cells derived cardiotoxicity testing platform for testing oncotherapeutic analogues of quercetin and cinnamic acid. Sci Rep 2022; 12:21362. [PMID: 36494370 PMCID: PMC9734143 DOI: 10.1038/s41598-022-21721-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022] Open
Abstract
Oncotherapeutics research is progressing at a rapid pace, however, not many drugs complete the successful clinical trial because of severe off-target toxicity to cardiomyocytes which ultimately leads to cardiac dysfunction. It is thus important to emphasize the need for early testing for possible cardiotoxicity of emerging oncotherapeutics. In this study, we assessed a novel stem cell-derived cardiac model for testing for cardiotoxicity of novel oncotherapeutics. We evaluated the cardiotoxic effect of synthesized derivatives of oncotherapeutics, quercetin (QMJ-2, -5, and -6) and cinnamic acid (NMJ-1, -2, and -3) using human Wharton's jelly mesenchymal stem cells-derived cardiomyocytes (WJCM) against known cardiotoxic oncologic drugs, doxorubicin, 5-fluorouracil, cisplatin. QMJ-6, NMJ-2, and NMJ-3 were not cardiotoxic and had minimum cardiac side effects. They did not show any effect on cardiomyocyte viability, caused low LDH release, and intracellular ROS production kept the calcium flux minimal and protected the active mitochondrial status in cardiomyocytes. They persevered cardiac-specific gene expression as well. However, compounds QMJ-2, QMJ-5, and NMJ-1 were cardiotoxic and the concentration needs to be reduced to prevent toxic effects on cardiomyocytes. Significantly, we were able to demonstrate that WJCM is an efficient cardiac testing model to analyze the cardiotoxicity of drugs in a human context.
Collapse
|
188
|
Mesenchymal stromal/stem cells: breaking the deadlock in the treatment of multiple organ dysfunction syndrome. Chin Med J (Engl) 2022; 135:2818-2820. [PMID: 36745767 PMCID: PMC9943974 DOI: 10.1097/cm9.0000000000002428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
189
|
Korpershoek JV, Rikkers M, Wallis FSA, Dijkstra K, te Raa M, de Knijff P, Saris DBF, Vonk LA. Mitochondrial Transport from Mesenchymal Stromal Cells to Chondrocytes Increases DNA Content and Proteoglycan Deposition In Vitro in 3D Cultures. Cartilage 2022; 13:133-147. [PMID: 36262105 PMCID: PMC9924973 DOI: 10.1177/19476035221126346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Allogeneic mesenchymal stromal cells (MSCs) are used in the 1-stage treatment of articular cartilage defects. The aim of this study is to investigate whether transport of mitochondria exists between chondrocytes and MSCs and to investigate whether the transfer of mitochondria to chondrocytes contributes to the mechanism of action of MSCs. DESIGN Chondrocytes and MSCs were stained with MitoTracker, and CellTrace was used to distinguish between cell types. The uptake of fluorescent mitochondria was measured in cocultures using flow cytometry. Transport was visualized using fluorescence microscopy. Microvesicles were isolated and the presence of mitochondria was assessed. Mitochondria were isolated from MSCs and transferred to chondrocytes using MitoCeption. Pellets of chondrocytes, chondrocytes with transferred MSC mitochondria, and cocultures were cultured for 28 days. DNA content and proteoglycan content were measured. Mitochondrial DNA of cultured pellets and of repair cartilage tissue was quantified. RESULTS Mitochondrial transfer occurred bidirectionally within the first 4 hours until 16 hours of coculture. Transport took place via tunneling nanotubes, direct cell-cell contact, and extracellular vesicles. After 28 days of pellet culture, DNA content and proteoglycan deposition were higher in chondrocyte pellets to which MSC mitochondria were transferred than the control groups. No donor mitochondrial DNA was traceable in the biopsies, whereas an increase in MSC mitochondrial DNA was seen in the pellets. CONCLUSIONS These results suggest that mitochondrial transport plays a role in the chondroinductive effect of MSCs on chondrocytes in vitro. However, in vivo no transferred mitochondria could be traced back after 1 year.
Collapse
Affiliation(s)
- Jasmijn V. Korpershoek
- UMC Utrecht, Utrecht, The
Netherlands,Daniel B. F. Saris, UMC Utrecht,
Huispostnummer G05.228, Heidelberglaan 100, Utrecht 3584 CX, The Netherlands.
| | | | | | | | - Marije te Raa
- Leids Universitair Medisch Centrum,
Leiden, The Netherlands
| | | | | | | |
Collapse
|
190
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
191
|
Scala P, Lovecchio J, Lamparelli EP, Vitolo R, Giudice V, Giordano E, Selleri C, Rehak L, Maffulli N, Della Porta G. Myogenic commitment of human stem cells by myoblasts Co-culture: a static vs. a dynamic approach. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:49-58. [PMID: 35188030 DOI: 10.1080/21691401.2022.2039684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An in-vitro model of human bone marrow mesenchymal stem cells (hBM-MSCs) myogenic commitment by synergic effect of a differentiation media coupled with human primary skeletal myoblasts (hSkMs) co-culture was developed adopting both conventional static co-seeding and perfused culture systems. Static co-seeding provided a notable outcome in terms of gene expression with a significant increase of Desmin (141-fold) and Myosin heavy chain II (MYH2, 32-fold) at day 21, clearly detected also by semi-quantitative immunofluorescence. Under perfusion conditions, myogenic induction ability of hSkMs on hBM-MSCs was exerted by paracrine effect with an excellent gene overexpression and immunofluorescence detection of MYH2 protein; furthermore, due to the dynamic cell culture in separate wells, western blot data were acquired confirming a successful cell commitment at day 14. A significant increase of anti-inflammatory cytokine gene expression, including IL-10 and IL-4 (15-fold and 11-fold, respectively) at day 14, with respect to the pro-inflammatory cytokines IL-12A (7-fold at day 21) and IL-1β (1.4-fold at day 7) was also detected during dynamic culture, confirming the immunomodulatory activity of hBM-MSCs along with commitment events. The present study opens interesting perspectives on the use of dynamic culture based on perfusion as a versatile tool to study myogenic events and paracrine cross-talk compared to the simple co-seeding static culture.
Collapse
Affiliation(s)
- Pasqualina Scala
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - J Lovecchio
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy
| | - E P Lamparelli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - R Vitolo
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - V Giudice
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - E Giordano
- Mol Cel Eng. Lab "S. Cavalcanti", Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Universitá 50, 47522 Cesena, Forlí-Cesena (FC), Italy.,Health Sciences and Technologies - Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano dell'Emilia, Bologna (BO), Italy.,Advanced Research Center on Electronic Systems (ARCES), University of Bologna, Via Vincenzo Toffano 2/2, 40125 Bologna (BO), Italy
| | - C Selleri
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - L Rehak
- Athena Biomedical innovations, Viale Europa 139, Florence (FI), 50126, Italy
| | - N Maffulli
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy
| | - G Della Porta
- Translational Medicine Laboratory, Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Salerno (SA), Italy.,Interdepartment Centre BIONAM, Università di Salerno, via Giovanni Paolo I, 84084 Fisciano, Salerno (SA), Italy
| |
Collapse
|
192
|
Robb KP, Audet J, Gandhi R, Viswanathan S. Putative critical quality attribute matrix identifies mesenchymal stromal cells with potent immunomodulatory and angiogenic "fitness" ranges in response to culture process parameters. Front Immunol 2022; 13:972095. [PMID: 36532069 PMCID: PMC9747767 DOI: 10.3389/fimmu.2022.972095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/27/2022] [Indexed: 12/05/2022] Open
Abstract
Adipose-derived mesenchymal stromal cells (MSC(AT)) display immunomodulatory and angiogenic properties, but an improved understanding of quantitative critical quality attributes (CQAs) that inform basal MSC(AT) fitness ranges for immunomodulatory and/or angiogenic applications is urgently needed for effective clinical translation. We constructed an in vitro matrix of multivariate readouts to identify putative CQAs that were sensitive enough to discriminate between specific critical processing parameters (CPPs) chosen for their ability to enhance MSC immunomodulatory and angiogenic potencies, with consideration for donor heterogeneity. We compared 3D aggregate culture conditions (3D normoxic, 3D-N) and 2D hypoxic (2D-H) culture as non-genetic CPP conditions that augment immunomodulatory and angiogenic fitness of MSC(AT). We measured multivariate panels of curated genes, soluble factors, and morphometric features for MSC(AT) cultured under varying CPP and licensing conditions, and we benchmarked these against two functional and therapeutically relevant anchor assays - in vitro monocyte/macrophage (MΦ) polarization and in vitro angiogenesis. Our results showed that varying CPP conditions was the primary driver of MSC(AT) immunomodulatory fitness; 3D-N conditions induced greater MSC(AT)-mediated MΦ polarization toward inflammation-resolving subtypes. In contrast, donor heterogeneity was the primary driver of MSC(AT) angiogenic fitness. Our analysis further revealed panels of putative CQAs with minimum and maximum values that consisted of twenty MSC(AT) characteristics that informed immunomodulatory fitness ranges, and ten MSC(AT) characteristics that informed angiogenic fitness ranges. Interestingly, many of the putative CQAs consisted of angiogenic genes or soluble factors that were inversely correlated with immunomodulatory functions (THBS1, CCN2, EDN1, PDGFA, VEGFA, EDIL3, ANGPT1, and ANG genes), and positively correlated to angiogenic functions (VEGF protein), respectively. We applied desirability analysis to empirically rank the putative CQAs for MSC(AT) under varying CPP conditions and donors to numerically identify the desirable CPP conditions or donors with maximal MSC(AT) immunomodulatory and/or angiogenic fitness. Taken together, our approach enabled combinatorial analysis of the matrix of multivariate readouts to provide putative quantitative CQAs that were sensitive to variations in select CPPs that enhance MSC immunomodulatory/angiogenic potency, and donor heterogeneity. These putative CQAs may be used to prospectively screen potent MSC(AT) donors or cell culture conditions to optimize for desired basal MSC(AT) immunomodulatory or angiogenic fitness.
Collapse
Affiliation(s)
- Kevin P. Robb
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Krembil Research Institute, University Health Network, Toronto, ON, Canada,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Julie Audet
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Rajiv Gandhi
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Department of Surgery, Division of Orthopedic Surgery, University of Toronto, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada,Krembil Research Institute, University Health Network, Toronto, ON, Canada,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada,Department of Medicine, Division of Hematology, University of Toronto, Toronto, ON, Canada,*Correspondence: Sowmya Viswanathan,
| |
Collapse
|
193
|
Alberti G, Russo E, Corrao S, Anzalone R, Kruzliak P, Miceli V, Conaldi PG, Di Gaudio F, La Rocca G. Current Perspectives on Adult Mesenchymal Stromal Cell-Derived Extracellular Vesicles: Biological Features and Clinical Indications. Biomedicines 2022; 10:2822. [PMID: 36359342 PMCID: PMC9687875 DOI: 10.3390/biomedicines10112822] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 08/10/2023] Open
Abstract
Extracellular vesicles (EVs) constitute one of the main mechanisms by which cells communicate with the surrounding tissue or at distance. Vesicle secretion is featured by most cell types, and adult mesenchymal stromal cells (MSCs) of different tissue origins have shown the ability to produce them. In recent years, several reports disclosed the molecular composition and suggested clinical indications for EVs derived from adult MSCs. The parental cells were already known for their roles in different disease settings in regulating inflammation, immune modulation, or transdifferentiation to promote cell repopulation. Interestingly, most reports also suggested that part of the properties of parental cells were maintained by isolated EV populations. This review analyzes the recent development in the field of cell-free therapies, focusing on several adult tissues as a source of MSC-derived EVs and the available clinical data from in vivo models.
Collapse
Affiliation(s)
- Giusi Alberti
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Eleonora Russo
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Simona Corrao
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Rita Anzalone
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Peter Kruzliak
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Vitale Miceli
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Pier Giulio Conaldi
- Research Department, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | | | - Giampiero La Rocca
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
194
|
iPSC-neural crest derived cells embedded in 3D printable bio-ink promote cranial bone defect repair. Sci Rep 2022; 12:18701. [PMID: 36333414 PMCID: PMC9636385 DOI: 10.1038/s41598-022-22502-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Cranial bone loss presents a major clinical challenge and new regenerative approaches to address craniofacial reconstruction are in great demand. Induced pluripotent stem cell (iPSC) differentiation is a powerful tool to generate mesenchymal stromal cells (MSCs). Prior research demonstrated the potential of bone marrow-derived MSCs (BM-MSCs) and iPSC-derived mesenchymal progenitor cells via the neural crest (NCC-MPCs) or mesodermal lineages (iMSCs) to be promising cell source for bone regeneration. Overexpression of human recombinant bone morphogenetic protein (BMP)6 efficiently stimulates bone formation. The study aimed to evaluate the potential of iPSC-derived cells via neural crest or mesoderm overexpressing BMP6 and embedded in 3D printable bio-ink to generate viable bone graft alternatives for cranial reconstruction. Cell viability, osteogenic potential of cells, and bio-ink (Ink-Bone or GelXa) combinations were investigated in vitro using bioluminescent imaging. The osteogenic potential of bio-ink-cell constructs were evaluated in osteogenic media or nucleofected with BMP6 using qRT-PCR and in vitro μCT. For in vivo testing, two 2 mm circular defects were created in the frontal and parietal bones of NOD/SCID mice and treated with Ink-Bone, Ink-Bone + BM-MSC-BMP6, Ink-Bone + iMSC-BMP6, Ink-Bone + iNCC-MPC-BMP6, or left untreated. For follow-up, µCT was performed at weeks 0, 4, and 8 weeks. At the time of sacrifice (week 8), histological and immunofluorescent analyses were performed. Both bio-inks supported cell survival and promoted osteogenic differentiation of iNCC-MPCs and BM-MSCs in vitro. At 4 weeks, cell viability of both BM-MSCs and iNCC-MPCs were increased in Ink-Bone compared to GelXA. The combination of Ink-Bone with iNCC-MPC-BMP6 resulted in an increased bone volume in the frontal bone compared to the other groups at 4 weeks post-surgery. At 8 weeks, both iNCC-MPC-BMP6 and iMSC-MSC-BMP6 resulted in an increased bone volume and partial bone bridging between the implant and host bone compared to the other groups. The results of this study show the potential of NCC-MPC-incorporated bio-ink to regenerate frontal cranial defects. Therefore, this bio-ink-cell combination should be further investigated for its therapeutic potential in large animal models with larger cranial defects, allowing for 3D printing of the cell-incorporated material.
Collapse
|
195
|
Tian C, Li Y, Wang L, Si J, Zheng Y, Kang J, Wang Y, You MJ, Zheng G. Blockade of FGF2/FGFR2 partially overcomes bone marrow mesenchymal stromal cells mediated progression of T-cell acute lymphoblastic leukaemia. Cell Death Dis 2022; 13:922. [PMID: 36333298 PMCID: PMC9636388 DOI: 10.1038/s41419-022-05377-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
The development of acute lymphoblastic leuakemia (ALL) is partly attributed to the effects of bone marrow (BM) microenvironment, especially mesenchymal stromal cells (MSCs), which interact bilaterally with leukaemia cells, leading to ALL progression. In order to find MSCs-based microenvironment targeted therapeutic strategies, Notch1-induced T-cell ALL (T-ALL) mice models were used and dynamic alterations of BM-MSCs with increased cell viability during T-ALL development was observed. In T-ALL mice derived stroma-based condition, leukaemia cells showed significantly elevated growth capacity indicating that MSCs participated in leukaemic niche formation. RNA sequence results revealed that T-ALL derived MSCs secreted fibroblast growth factor 2 (FGF2), which combined with fibroblast growth factor receptor 2 (FGFR2) on leukaemia cells, resulting in activation of PI3K/AKT/mTOR signalling pathway in leukaemia cells. In vitro blocking the interaction between FGF2 and FGFR2 with BGJ398 (infigratinib), a FGFR1-3 kinase inhibitor, or knockdown FGF2 in MSCs by interference caused deactivation of PI3K/AKT/mTOR pathway and dysregulations of genes associated with cell cycle and apoptosis in ALL cells, leading to decrease of leukaemia cells. In mouse model received BGJ398, overall survival was extended and dissemination of leukaemia cells in BM, spleen, liver and peripheral blood was decreased. After subcutaneous injection of primary human T-ALL cells with MSCs, tumour growth was suppressed when FGF2/FGFR2 was interrupted. Thus, inhibition of FGF2/FGFR2 interaction appears to be a valid strategy to overcome BM-MSCs mediated progression of T-ALL, and BGJ398 could indeed improve outcomes in T-ALL, which provide theoretical basis of BGJ398 as a BM microenvironment based therapeutic strategy to control disease progression.
Collapse
Affiliation(s)
- Chen Tian
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Yueyang Li
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Lina Wang
- grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Junqi Si
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Yaxin Zheng
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Junnan Kang
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| | - Yafei Wang
- grid.411918.40000 0004 1798 6427Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, 300060 China
| | - M. James You
- grid.240145.60000 0001 2291 4776Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77479 USA
| | - Guoguang Zheng
- grid.506261.60000 0001 0706 7839State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020 China
| |
Collapse
|
196
|
Akasaka Y. The Role of Mesenchymal Stromal Cells in Tissue Repair and Fibrosis. Adv Wound Care (New Rochelle) 2022; 11:561-574. [PMID: 34841889 DOI: 10.1089/wound.2021.0037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Significance: The present review covers an overview of the current understanding of biology of mesenchymal stromal cells (MSCs) and suggests an important role of their differential potential for clinical approaches associated with tissue repair and fibrosis. Recent Advances: Genetic lineage tracing technology has enabled the delineation of cellular hierarchies and examination of MSC cellular origins and myofibroblast sources. This technique has led to the characterization of perivascular MSC populations and suggests that pericytes might provide a local source of tissue-specific MSCs, which can differentiate into tissue-specific cells for tissue repair and fibrosis. Autologous adipose tissue MSCs led to the advance in tissue engineering for regeneration of damaged tissues. Critical Issues: Recent investigation has revealed that perivascular MSCs might be the origin of myofibroblasts during fibrosis development, and perivascular MSCs might be the major source of myofibroblasts in fibrogenic disease. Adipose tissue MSCs combined with cytokines and biomaterials are available in the treatment of soft tissue defect and skin wound healing. Future Directions: Further investigation of the roles of perivascular MSCs may enable new approaches in the treatment of fibrogenic disease; moreover, perivascular MSCs might have potential as an antifibrotic target for fibrogenic disease. Autologous adipose tissue MSCs combined with cytokines and biomaterials will be an alternative method for the treatment of soft tissue defect and skin wound healing.
Collapse
Affiliation(s)
- Yoshikiyo Akasaka
- Division of Research Promotion and Development, Advanced Research Center, Toho University Graduate School of Medicine, Ota-ku, Japan.,Department of Pathology, Toho University School of Medicine, Ota-ku, Japan
| |
Collapse
|
197
|
Zhao Z, Behm C, Tian Z, Rausch MA, Rausch-Fan X, Andrukhov O. Cyclic tensile strain-induced yes-associated protein activity modulates the response of human periodontal ligament mesenchymal stromal cells to tumor necrosis factor-α. Arch Oral Biol 2022; 143:105527. [DOI: 10.1016/j.archoralbio.2022.105527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/02/2022]
|
198
|
Subhan BS, Ki M, Verzella A, Shankar S, Rabbani PS. Behind the Scenes of Extracellular Vesicle Therapy for Skin Injuries and Disorders. Adv Wound Care (New Rochelle) 2022; 11:575-597. [PMID: 34806432 PMCID: PMC9419953 DOI: 10.1089/wound.2021.0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 11/10/2021] [Indexed: 01/29/2023] Open
Abstract
Significance: Skin wounds and disorders compromise the protective functions of skin and patient quality of life. Although accessible on the surface, they are challenging to address due to paucity of effective therapies. Exogenous extracellular vesicles (EVs) and cell-free derivatives of adult multipotent stromal cells (MSCs) are developing as a treatment modality. Knowledge of origin MSCs, EV processing, and mode of action is necessary for directed use of EVs in preclinical studies and methodical translation. Recent Advances: Nanoscale to microscale EVs, although from nonskin cells, induce functional responses in cutaneous wound cellular milieu. EVs allow a shift from cell-based to cell-free/derived modalities by carrying the MSC beneficial factors but eliminating risks associated with MSC transplantation. EVs have demonstrated striking efficacy in resolution of preclinical wound models, specifically within the complexity of skin structure and wound pathology. Critical Issues: To facilitate comparison across studies, tissue sources and processing of MSCs, culture conditions, isolation and preparations of EVs, and vesicle sizes require standardization as these criteria influence EV types and contents, and potentially determine the induced biological responses. Procedural parameters for all steps preceding the actual therapeutic administration may be the key to generating EVs that demonstrate consistent efficacy through known mechanisms. We provide a comprehensive review of such parameters and the subsequent tissue, cellular and molecular impact of the derived EVs in different skin wounds/disorders. Future Directions: We will gain more complete knowledge of EV-induced effects in skin, and specificity for different wounds/conditions. The safety and efficacy of current preclinical xenogenic applications will favor translation into allogenic clinical applications of EVs as a biologic.
Collapse
Affiliation(s)
- Bibi S. Subhan
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Michelle Ki
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Alexandra Verzella
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Shruthi Shankar
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| | - Piul S. Rabbani
- Hansjörg Wyss Department of Plastic Surgery, New York University School of Medicine, New York, New York, USA
| |
Collapse
|
199
|
Wang Y, Fang J, Liu B, Shao C, Shi Y. Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell 2022; 29:1515-1530. [DOI: 10.1016/j.stem.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
|
200
|
Ho KKW, Lee WYW, Griffith JF, Ong MTY, Li G. Randomized control trial of mesenchymal stem cells versus hyaluronic acid in patients with knee osteoarthritis - A Hong Kong pilot study. J Orthop Translat 2022; 37:69-77. [PMID: 36262962 PMCID: PMC9550852 DOI: 10.1016/j.jot.2022.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/11/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Objective This pilot study evaluated the efficacy of autologous bone marrow-derived mesenchymal stem cells (BM-MSCs) versus hyaluronic acid (HA) in surgically naïve patients with knee osteoarthritis (OA). Methods Single-centre, single-blind randomized study of patients with knee OA. Twenty patients were randomized into groups of 10 each for intra-articular injection of cultured BM-MSCs (6 ml of BM-MSCs at 1 × 106 cells/mL) or HA (6 ml). Clinical assessments of pain, quality of life, radiographic imaging, and magnetic resonance imaging (MRI) compositional change were performed at baseline and 12 months follow-up. Results Compared with HA, BM-MSCs injection resulted in significant improvement in qualify of life and reduction in pain as reflected by visual analogue scale (VAS) pain score, Western Ontario and McMaster Universities Arthritis Index (WOMAC) score, and 36-Item Short Form Survey (SF-36) score collectively. T2-relaxation time tended to decrease more in the BM-MSCs group with a 38 ± 24.0% reduction in 6 out of 10 BM-MSC participants; while there was only a 12 ± 7.9% reduction in 4 out of 10 HA participants at the end of follow-up. The remaining participants showed either no response or had relaxation time increased on MRI assessment. Conclusions This pilot study found that autologous BM-MSCs significantly reduced pain, improved functional assessment score, and improved quality of life parameters comparing with HA at one year follow-up. Further clinical trial with larger sample size and longer follow up duration is warranted. The Translational Potential of this Article This pilot RCT demonstrated the feasibility and potential effectiveness of BM-MSCs advanced therapy for patients with knee OA compared to HA injection. Further multi-center clinical trial with a larger sample size and longer follow up duration in accordance with latest regulatory guidelines is warranted to ascertain the long term safety and effectiveness of MSCs therapy for cartilage regeneration in OA. Registration The study was registered in the Centre for Clinical Research Biostatistics - Clinical Trials Registry (CUHK_CCT00469).
Collapse
Affiliation(s)
- Kevin Ki-Wai Ho
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Wayne Yuk-Wai Lee
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - James F Griffith
- Department of Imaging and Interventional Radiology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Michael Tim-Yun Ong
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.,Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| |
Collapse
|