1
|
Lopes-Pacheco M, Rocco PRM. Functional enhancement strategies to potentiate the therapeutic properties of mesenchymal stromal cells for respiratory diseases. Front Pharmacol 2023; 14:1067422. [PMID: 37007034 PMCID: PMC10062457 DOI: 10.3389/fphar.2023.1067422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Respiratory diseases remain a major health concern worldwide because they subject patients to considerable financial and psychosocial burdens and result in a high rate of morbidity and mortality. Although significant progress has been made in understanding the underlying pathologic mechanisms of severe respiratory diseases, most therapies are supportive, aiming to mitigate symptoms and slow down their progressive course but cannot improve lung function or reverse tissue remodeling. Mesenchymal stromal cells (MSCs) are at the forefront of the regenerative medicine field due to their unique biomedical potential in promoting immunomodulation, anti-inflammatory, anti-apoptotic and antimicrobial activities, and tissue repair in various experimental models. However, despite several years of preclinical research on MSCs, therapeutic outcomes have fallen far short in early-stage clinical trials for respiratory diseases. This limited efficacy has been associated with several factors, such as reduced MSC homing, survival, and infusion in the late course of lung disease. Accordingly, genetic engineering and preconditioning methods have emerged as functional enhancement strategies to potentiate the therapeutic actions of MSCs and thus achieve better clinical outcomes. This narrative review describes various strategies that have been investigated in the experimental setting to functionally potentiate the therapeutic properties of MSCs for respiratory diseases. These include changes in culture conditions, exposure of MSCs to inflammatory environments, pharmacological agents or other substances, and genetic manipulation for enhanced and sustained expression of genes of interest. Future directions and challenges in efficiently translating MSC research into clinical practice are discussed.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Miquéias Lopes-Pacheco, ; Patricia R. M. Rocco,
| |
Collapse
|
2
|
Muacevic A, Adler JR. Congenital Pseudoarthrosis of the Tibia: A Narrative Review. Cureus 2022; 14:e32501. [PMID: 36654595 PMCID: PMC9840411 DOI: 10.7759/cureus.32501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022] Open
Abstract
Congenital pseudoarthrosis of the tibia, also known as CPT, is a rare bone disease mostly occurring in the pediatric age group of 10. The case generally represents anterolateral bowing of the tibia with refractures. This condition is quite rare and has very few cases per year. CPT is still a challenging concern in orthopedics. Bone reunion often requires repeated surgical measurements. With advancements, we have new treatment plans that include induced membrane techniques and bone marrow stromal cell grafts of various induced and morphogenetic protein, which still requires confirmation. Different treatment strategies based on surgical, mechanical, and biological concepts have been shown with varying success rates. Ilizarov technique and vascularised fibular grafts have vastly increased the effectiveness in treating CPT of the tibia. Constant and recurrent refracture, residual deformities, and difficulty obtaining bone union remain the challenges in managing CPT. Hence, phasing CPT as bowing the tibia in an anterolateral fashion is more appropriate because it's a heterogeneous entity with varying prognoses.
Collapse
|
3
|
Hastreiter AA, Dos Santos GG, Makiyama EN, Santos EWC, Borelli P, Fock RA. Effects of protein malnutrition on hematopoietic regulatory activity of bone marrow mesenchymal stem cells. J Nutr Biochem 2021; 93:108626. [PMID: 33705953 DOI: 10.1016/j.jnutbio.2021.108626] [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: 12/16/2019] [Revised: 12/15/2020] [Accepted: 02/02/2021] [Indexed: 11/29/2022]
Abstract
Protein malnutrition causes anemia and leukopenia as it reduces hematopoietic precursors and impairs the production of mediators that regulate hematopoiesis. Hematopoiesis occurs in distinct bone marrow niches that modulate the processes of differentiation, proliferation and self-renewal of hematopoietic stem cells (HSCs). Mesenchymal stem cells (MSCs) contribute to the biochemical composition of bone marrow niches by the secretion of several growth factors and cytokines, and they play an important role in the regulation of HSCs and hematopoietic progenitors. In this study, we investigated the effect of protein malnutrition on the hematopoietic regulatory function of MSCs. C57BL/6NTaq mice were divided into control and protein malnutrition groups, which received, respectively, a normal protein diet (12% casein) and a low protein diet (2% casein). The results showed that protein malnutrition altered the synthesis of SCF, TFG-β, Angpt-1, CXCL-12, and G-CSF by MSCs. Additionally, MSCs from the protein malnutrition group were not able to maintain the lymphoid, granulocytic and megakaryocytic-erythroid differentiation capacity compared to the MSCs of the control group. In this way, the comprehension of the role of MSCs on the regulation of the hematopoietic cells, in protein malnutrition states, is for the first time showed. Therefore, we infer that hematopoietic alterations caused by protein malnutrition are due to multifactorial alterations and, at least in part, the MSCs' contribution to hematological impairment.
Collapse
Affiliation(s)
- Araceli Aparecida Hastreiter
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Guilherme G Dos Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edson Naoto Makiyama
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ed Wilson Cavalcante Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Primavera Borelli
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
4
|
de Souza BM, Rodrigues M, de Oliveira FS, da Silva LPA, Bouças AP, Portinho CP, Dos Santos BP, Camassola M, Rocha D, Lysakowski S, Martini J, Leitão CB, Nardi NB, Bauer AC, Crispim D. Improvement of human pancreatic islet quality after co-culture with human adipose-derived stem cells. Mol Cell Endocrinol 2020; 505:110729. [PMID: 31972330 DOI: 10.1016/j.mce.2020.110729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/30/2019] [Accepted: 01/17/2020] [Indexed: 01/08/2023]
Abstract
The aim of this study was to investigate whether co-culture of human islets with adipose-derived stem cells (ASCs) can improve islet quality and to evaluate which factors play a role in the protective effect of ASCs against islet dysfunction. Islets and ASCs were cultured in three experimental groups for 24 h, 48 h, and 72 h: 1) indirect co-culture of islets with ASC monolayer (Islets/ASCs); 2) islets alone; and 3) ASCs alone. Co-culture with ASCs improved islet viability and function in all culture time-points analyzed. VEGFA, HGF, IL6, IL8, IL10, CCL2, IL1B, and TNF protein levels were increased in supernatants of islet/ASC group compared to islets alone, mainly after 24 h. Moreover, VEGFA, IL6, CCL2, HIF1A, XIAP, CHOP, and NFKBIA genes were differentially expressed in islets from the co-culture condition compared to islets alone. In conclusion, co-culture of islets with ASCs promotes improvements in islet quality.
Collapse
Affiliation(s)
- Bianca M de Souza
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil.
| | - Michelle Rodrigues
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Fernanda S de Oliveira
- Laboratory of Cell Differentiation, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Liana P A da Silva
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Ana P Bouças
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Ciro P Portinho
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil
| | - Bruno P Dos Santos
- Laboratory for Tissue Bioengineering (BioTis), Inserm U1026, University of Bordeaux, Bordeaux, France
| | - Melissa Camassola
- Laboratory for Stem Cells and Tissue Engineering, Post-Graduation Program in Cellular and Molecular Biology Applied to Health, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Dagoberto Rocha
- Post-Graduation Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
| | - Simone Lysakowski
- Organ Procurement Organization, Santa Casa de Misericórdia de Porto Alegre. Porto Alegre, RS, Brazil
| | - Juliano Martini
- Transplant Center, Surgery Department, Hospital Dom Vicente Scherer, Santa Casa de Misericórdia de Porto Alegre. Porto Alegre, RS, Brazil
| | - Cristiane B Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Nance B Nardi
- Laboratory for Stem Cells and Tissue Engineering, Post-Graduation Program in Cellular and Molecular Biology Applied to Health, Universidade Luterana do Brasil, Canoas, RS, Brazil
| | - Andrea C Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clınicas de Porto Alegre, Porto Alegre, Rio Grande do Sul (RS), Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, RS, Brazil
| |
Collapse
|
5
|
Ni H, Zhao Y, Ji Y, Shen J, Xiang M, Xie Y. Adipose-derived stem cells contribute to cardiovascular remodeling. Aging (Albany NY) 2019; 11:11756-11769. [PMID: 31800397 PMCID: PMC6932876 DOI: 10.18632/aging.102491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/17/2019] [Indexed: 02/06/2023]
Abstract
Obesity is an independent risk factor for cardiovascular disease. Adipose tissue was initially thought to be involved in metabolism through paracrine. Recent researches discovered mesenchymal stem cells inside adipose tissue which could differentiate into vascular lineages in vitro and in vivo, participating vascular remodeling. However, there were few researches focusing on distinct characteristics and functions of adipose-derived stem cells (ADSCs) from different regions. This is the first comprehensive review demonstrating the variances of ADSCs from the perspective of their origins.
Collapse
Affiliation(s)
- Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
He X, Yang Y, Yao MW, Ren TT, Guo W, Li L, Xu X. Full title: High glucose protects mesenchymal stem cells from metformin-induced apoptosis through the AMPK-mediated mTOR pathway. Sci Rep 2019; 9:17764. [PMID: 31780804 PMCID: PMC6882892 DOI: 10.1038/s41598-019-54291-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 09/02/2019] [Indexed: 12/14/2022] Open
Abstract
Micro- and macro-vascular events are directly associated with hyperglycemia in patients with type 2 diabetes mellitus (T2DM), but whether intensive glucose control decreases the risk of diabetic cardiovascular complications remains uncertain. Many studies have confirmed that impaired quality and quantity of mesenchymal stem cells (MSCs) plays a pathogenic role in diabetes. Our previous study found that the abundance of circulating MSCs was significantly decreased in patients with T2DM, which was correlated with the progression of diabetic complications. In addition, metformin-induced MSC apoptosis is one of the reasons for the decreased quantity of endogenous or exogenous MSCs during intensive glucose control. However, the role of glucose in metformin-induced MSC apoptosis during intensive glucose control in T2DM remains unknown. In this study, we found that metformin induces MSC apoptosis during intensive glucose control, while high glucose (standard glucose control) could significantly reverse its adverse effect in an AMPK-mTOR pathway dependent manner. Thus, our results indicate that the poorer clinical benefit of the intensive glucose control strategy may be related to an adverse effect due to metformin-induced MSC apoptosis during intensive glucose control therapy in patients with T2DM.
Collapse
Affiliation(s)
- Xiao He
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- PLA Rocket Force Characteristic Medical Center, Beijing, P.R. China
| | - Yi Yang
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department of Rheumatology and Clinical Immunology, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
| | - Meng-Wei Yao
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, P.R. China
| | - Ting-Ting Ren
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Department of Histology and Embryology, Qingdao University Medical College, Qingdao, Shandong, P.R. China
| | - Wei Guo
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China
| | - Ling Li
- Department of Histology and Embryology, Qingdao University Medical College, Qingdao, Shandong, P.R. China
| | - Xiang Xu
- Department of Stem Cell and Regenerative Medicine, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China.
- Central Laboratory, State Key laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Military Medical University, Chongqing, P.R. China.
- Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Army Medical University, Chongqing, P.R. China.
| |
Collapse
|
7
|
Lopes-Pacheco M, Robba C, Rocco PRM, Pelosi P. Current understanding of the therapeutic benefits of mesenchymal stem cells in acute respiratory distress syndrome. Cell Biol Toxicol 2019; 36:83-102. [PMID: 31485828 PMCID: PMC7222160 DOI: 10.1007/s10565-019-09493-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 08/27/2019] [Indexed: 12/20/2022]
Abstract
The acute respiratory distress syndrome (ARDS) is a multifaceted lung disorder in which no specific therapeutic intervention is able to effectively improve clinical outcomes. Despite an improved understanding of molecular mechanisms and advances in supportive care strategies, ARDS remains associated with high mortality, and survivors usually face long-term morbidity. In recent years, preclinical studies have provided mounting evidence of the potential of mesenchymal stem cell (MSC)-based therapies in lung diseases and critical illnesses. In several models of ARDS, MSCs have been demonstrated to induce anti-inflammatory and anti-apoptotic effects, improve epithelial and endothelial cell recovery, and enhance microbial and alveolar fluid clearance, thus resulting in improved lung and distal organ function and survival. Early-stage clinical trials have also demonstrated the safety of MSC administration in patients with ARDS, but further, large-scale investigations are required to assess the safety and efficacy profile of these therapies. In this review, we summarize the main mechanisms whereby MSCs have been shown to exert therapeutic effects in experimental ARDS. We also highlight questions that need to be further elucidated and barriers that must be overcome in order to efficiently translate MSC research into clinical practice.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Patricia Rieken Macêdo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy. .,Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy.
| |
Collapse
|
8
|
Ho YT, Shimbo T, Wijaya E, Ouchi Y, Takaki E, Yamamoto R, Kikuchi Y, Kaneda Y, Tamai K. Chromatin accessibility identifies diversity in mesenchymal stem cells from different tissue origins. Sci Rep 2018; 8:17765. [PMID: 30531792 PMCID: PMC6288149 DOI: 10.1038/s41598-018-36057-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/14/2018] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which can differentiate into tri-lineage (osteoblast, adipocyte, and chondrocyte) and suppress inflammation, are promising tools for regenerative medicine. MSCs are phenotypically diverse based on their tissue origins. However, the mechanisms underlying cell-type-specific gene expression patterns are not fully understood due to the lack of suitable strategy to identify the diversity. In this study, we investigated gene expression programs and chromatin accessibilities of MSCs by whole-transcriptome RNA-seq analysis and an assay for transposase-accessible chromatin using sequencing (ATAC-seq). We isolated MSCs from four tissues (femoral and vertebral bone marrow, adipose tissue, and lung) and analysed their molecular signatures. RNA-seq identified the expression of MSC markers and both RNA-seq and ATAC-seq successfully clustered the MSCs based on their tissue origins. Interestingly, clustering based on tissue origin was more accurate with chromatin accessibility signatures than with transcriptome profiles. Furthermore, we identified transcription factors potentially involved in establishing cell-type specific chromatin structures. Thus, epigenome analysis is useful to analyse MSC identity and can be utilized to characterize these cells for clinical use.
Collapse
Affiliation(s)
- Yen-Ting Ho
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takashi Shimbo
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Edward Wijaya
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,StemRIM Co., Ltd., Ibaraki, Osaka, Japan
| | - Yuya Ouchi
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,StemRIM Co., Ltd., Ibaraki, Osaka, Japan
| | - Eiichi Takaki
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,StemRIM Co., Ltd., Ibaraki, Osaka, Japan
| | - Ryoma Yamamoto
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.,StemRIM Co., Ltd., Ibaraki, Osaka, Japan
| | - Yasushi Kikuchi
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
9
|
Dilogo IH, Mujadid F, Nurhayati RW, Kurniawan A. Evaluation of bone marrow-derived mesenchymal stem cell quality from patients with congenital pseudoarthrosis of the tibia. J Orthop Surg Res 2018; 13:266. [PMID: 30352605 PMCID: PMC6199809 DOI: 10.1186/s13018-018-0977-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 10/17/2018] [Indexed: 02/18/2023] Open
Abstract
Background The treatment of congenital pseudoarthrosis of the tibia (CPT) remains challenging in pediatric orthopedics due to the difficulties in bone union, continuous angulation, joint stiffness, and severe limb length discrepancy. Mesenchymal stem cells (MSCs) therapy offers a complementary approach to improve the conventional surgical treatments. Although the autologous MSC treatment shows a promising strategy to promote bone healing in CPT patients, the quality of MSCs from CPT patients has not been well studied. The purpose of this study is to investigate the quality of MSCs isolated from patients with CPT. Methods The bone marrow-derived MSCs from the fracture site and iliac crest of six CPT patients were isolated and compared. The cumulative population doubling level (cPDL), phenotype characteristics, and trilineage differentiation potency were observed to assess the quality of both MSCs. Results There were no significant differences of the MSCs derived from the fracture site and the MSCs from the iliac crest of the subjects, in terms of cPDL, phenotype characteristics, and trilineage differentiation potency (all p > 0.05). However, MSCs from the fracture site had a higher senescence tendency than those from the iliac crest. Conclusion MSC quality is not the main reason for delayed bone regeneration in those with CPT. Thus, autologous MSC is a promising source for treating CPT patients
Collapse
Affiliation(s)
- Ismail Hadisoebroto Dilogo
- Integrated Service Unit of Stem Cell Medical Technology, Dr. Cipto Mangunkusumo General Hospital (RSCM), Jl. Diponegoro No 71, Salemba, Cental Jakarta, 10430, Indonesia. .,Stem Cell and Tissue Engineering Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No 6, Salemba, Cental Jakarta, 10430, Indonesia. .,Department of Orthopaedic and Traumatology, Faculty of Medicine, Universitas Indonesia - Dr. Cipto Mangunkusumo General Hospital, Jl. Diponegoro No 71, Salemba, Cental Jakarta, 10430, Indonesia.
| | - Fajar Mujadid
- Integrated Service Unit of Stem Cell Medical Technology, Dr. Cipto Mangunkusumo General Hospital (RSCM), Jl. Diponegoro No 71, Salemba, Cental Jakarta, 10430, Indonesia
| | - Retno Wahyu Nurhayati
- Stem Cell and Tissue Engineering Cluster, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No 6, Salemba, Cental Jakarta, 10430, Indonesia.,Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya No. 6, Central Jakarta, 10430, Indonesia
| | - Aryadi Kurniawan
- Department of Orthopaedic and Traumatology, Faculty of Medicine, Universitas Indonesia - Dr. Cipto Mangunkusumo General Hospital, Jl. Diponegoro No 71, Salemba, Cental Jakarta, 10430, Indonesia
| |
Collapse
|
10
|
de Mendonça L, Felix NS, Blanco NG, Da Silva JS, Ferreira TP, Abreu SC, Cruz FF, Rocha N, Silva PM, Martins V, Capelozzi VL, Zapata-Sudo G, Rocco PRM, Silva PL. Mesenchymal stromal cell therapy reduces lung inflammation and vascular remodeling and improves hemodynamics in experimental pulmonary arterial hypertension. Stem Cell Res Ther 2017; 8:220. [PMID: 28974252 PMCID: PMC5627397 DOI: 10.1186/s13287-017-0669-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
Background Experimental research has reported beneficial effects of mesenchymal stromal cell (MSC) therapy in pulmonary arterial hypertension (PAH). However, these studies either were based on prophylactic protocols or assessed basic remodeling features without evaluating possible mechanisms. We analyzed the effects of MSC therapy on lung vascular remodeling and hemodynamics and its possible mechanisms of action in monocrotaline (MCT)-induced PAH. Methods Twenty-eight Wistar rats were randomly divided into two groups. In the PAH group, animals received MCT 60 mg/kg intraperitoneally, while a control group received saline (SAL) instead. On day 14, both groups were further randomized to receive 105 adipose-derived MSCs or SAL intravenously (n = 7/group). On day 28, right ventricular systolic pressure (RVSP) and the gene expression of mediators associated with apoptosis, inflammation, fibrosis, Smad-1 levels, cell proliferation, and endothelial–mesenchymal transition were determined. In addition, lung histology (smooth muscle cell proliferation and plexiform-like injuries), CD68+ and CD163+ macrophages, and plasma levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) were evaluated. Results In the PAH group, adipose-derived MSCs, compared to SAL, reduced mean RVSP (29 ± 1 vs 39 ± 2 mmHg, p < 0.001), lung tissue collagen fiber content, smooth muscle cell proliferation, CD68+ macrophages, interleukin-6 expression, and the antiapoptotic mediators Bcl-2 and survivin. Conversely, expression of the proapoptotic mediator procaspase-3 and plasma VEGF increased, with no changes in PDGF. In the pulmonary artery, MSCs dampened the endothelial–mesenchymal transition. Conclusion In MCT-induced PAH, MSC therapy reduced lung vascular remodeling, thus improving hemodynamics. These beneficial effects were associated with increased levels of proapoptotic markers, mesenchymal-to-endothelial transition, reduced cell proliferation markers, and inflammation due to a shift away from the M1 phenotype. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0669-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lucas de Mendonça
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nathane S Felix
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Natália G Blanco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Jaqueline S Da Silva
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Tatiana P Ferreira
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Nazareth Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Physiology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Patrícia M Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute-Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Vanessa Martins
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Vera L Capelozzi
- Laboratory of Histomorphometry and Lung Genomics, University of São Paulo Faculty of Medicine, São Paulo, SP, Brazil
| | - Gizele Zapata-Sudo
- Laboratory of Cardiovascular Pharmacology, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
11
|
Pillai ICL, Li S, Romay M, Lam L, Lu Y, Huang J, Dillard N, Zemanova M, Rubbi L, Wang Y, Lee J, Xia M, Liang O, Xie YH, Pellegrini M, Lusis AJ, Deb A. Cardiac Fibroblasts Adopt Osteogenic Fates and Can Be Targeted to Attenuate Pathological Heart Calcification. Cell Stem Cell 2016; 20:218-232.e5. [PMID: 27867037 DOI: 10.1016/j.stem.2016.10.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 08/11/2016] [Accepted: 10/12/2016] [Indexed: 02/07/2023]
Abstract
Mammalian tissues calcify with age and injury. Analogous to bone formation, osteogenic cells are thought to be recruited to the affected tissue and induce mineralization. In the heart, calcification of cardiac muscle leads to conduction system disturbances and is one of the most common pathologies underlying heart blocks. However the cell identity and mechanisms contributing to pathological heart muscle calcification remain unknown. Using lineage tracing, murine models of heart calcification and in vivo transplantation assays, we show that cardiac fibroblasts (CFs) adopt an osteoblast cell-like fate and contribute directly to heart muscle calcification. Small-molecule inhibition of ENPP1, an enzyme that is induced upon injury and regulates bone mineralization, significantly attenuated cardiac calcification. Inhibitors of bone mineralization completely prevented ectopic cardiac calcification and improved post injury heart function. Taken together, these findings highlight the plasticity of fibroblasts in contributing to ectopic calcification and identify pharmacological targets for therapeutic development.
Collapse
Affiliation(s)
- Indulekha C L Pillai
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Shen Li
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Milagros Romay
- Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, UCLA, CA 90095, USA
| | - Larry Lam
- Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Yan Lu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Jie Huang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Nathaniel Dillard
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Marketa Zemanova
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Liudmilla Rubbi
- Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Yibin Wang
- Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Anesthesiology, UCLA, CA 90095, USA; Department of Physiology, UCLA, CA 90095, USA
| | - Jason Lee
- Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine and Crump Institute for Molecular Imaging, UCLA, CA 90095, USA
| | - Ming Xia
- Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA; Department of Materials Science & Engineering, School of Engineering, UCLA, CA 90095, USA
| | - Owen Liang
- Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA; Department of Materials Science & Engineering, School of Engineering, UCLA, CA 90095, USA
| | - Ya-Hong Xie
- Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA; Department of Materials Science & Engineering, School of Engineering, UCLA, CA 90095, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, UCLA, CA 90095, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), CA 90095, USA; Cardiovascular Research Laboratory, David Geffen School of Medicine, UCLA, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, School of Letters and Sciences, UCLA, CA 90095, USA; Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, CA 90095, USA; Molecular Biology Institute, UCLA, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, CA 90095, USA.
| |
Collapse
|
12
|
Santos BPD, da Costa Diesel LF, da Silva Meirelles L, Nardi NB, Camassola M. Identification of suitable reference genes for quantitative gene expression analysis in rat adipose stromal cells induced to trilineage differentiation. Gene 2016; 594:211-219. [PMID: 27601259 DOI: 10.1016/j.gene.2016.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/03/2016] [Accepted: 09/02/2016] [Indexed: 10/21/2022]
Abstract
This study was designed to (i) identify stable reference genes for the analysis of gene expression during in vitro differentiation of rat adipose stromal cells (rASCs), (ii) recommend stable genes for individual treatment conditions, and (iii) validate these genes by comparison with normalization results from stable and unstable reference genes. On the basis of a literature review, eight genes were selected: Actb, B2m, Hprt1, Ppia, Rplp0, Rpl13a, Rpl5, and Ywhaz. Genes were ranked according to their stability under different culture conditions as assessed using GenNorm, NormFinder, and RefFinder algorithms. Although the employed algorithms returned different rankings, the most frequently top-ranked genes were: B2m and/or Ppia for all 28day treatments (ALL28); Ppia and Hprt1 (adipogenic differentiation; A28), B2m (chondrogenic differentiation; C28), Rpl5 (controls maintained in complete culture medium; CCM), Rplp0 (osteogenic differentiation for 3days; O3), Rpl13a and Actb (osteogenic differentiation for 7days; O7), Rplp0 and Ppia (osteogenic differentiation for 14days; O14), Hprt1 and Ppia (osteogenic differentiation for 28days; O28), as well as Actb (all osteogenesis time points combined; ALLOSTEO). The obtained results indicate that the performance of reference genes depends on the differentiation protocol and on the analysis time, thus providing valuable information for the design of RT-PCR experiments.
Collapse
Affiliation(s)
- Bruno Paiva Dos Santos
- Laboratory of Stem Cells and Tissue Engineering, Lutheran University of Brazil, Canoas 92425-900, RS, Brazil.
| | | | - Lindolfo da Silva Meirelles
- Laboratory of Stem Cells and Tissue Engineering, Lutheran University of Brazil, Canoas 92425-900, RS, Brazil.
| | - Nance Beyer Nardi
- Laboratory of Stem Cells and Tissue Engineering, Lutheran University of Brazil, Canoas 92425-900, RS, Brazil.
| | - Melissa Camassola
- Laboratory of Stem Cells and Tissue Engineering, Lutheran University of Brazil, Canoas 92425-900, RS, Brazil.
| |
Collapse
|
13
|
Onzi GR, Ledur PF, Hainzenreder LD, Bertoni APS, Silva AO, Lenz G, Wink MR. Analysis of the safety of mesenchymal stromal cells secretome for glioblastoma treatment. Cytotherapy 2016; 18:828-37. [PMID: 27210718 DOI: 10.1016/j.jcyt.2016.03.299] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 03/05/2016] [Accepted: 03/31/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND AIMS The purpose of this study was to investigate whether the secretome of human adipose-derived stem cells (hADSC) affects human glioblastoma (GBM) cancer stem cell (CSC) subpopulation or has any influence on drug resistance and cell migration, evaluating the safety of hADSCs for novel cancer therapies. METHODS hADSCs were maintained in contact with fresh culture medium to produce hADSCs conditioned medium (CM). GBM U87 cells were cultured with CM and sphere formation, expression of genes related to resistance and CSCs-MGMT, OCT4, SOX2, NOTCH1, MSI1-and protein expression of OCT4 and Nanog were analyzed. The influence of hADSC CM on GBM resistance to temozolomide (TMZ) was evaluated by measuring cumulative population doubling and hADSC CM influence on tumor cell migration was analyzed using transwell assay. RESULTS hADSC CM did not alter CSC-related features such as sphere-forming capacity and expression of genes related to CSC. hADSC CM treatment alone did not change proliferation rate of U87 cells and, most important, did not alter the response of tumor cells to TMZ. However, hADSC CM secretome increased the migration capacity of glioblastoma cells. DISCUSSION hADSC CM neither induced an enrichment of CSCs in U87 cells population nor interfered in the response to TMZ in culture. Nevertheless, paracrine factors released by hADSCs were able to modulate glioblastoma cells migration. These findings provide novel information regarding the safety of using hADSCs against cancer and highlight the importance of considering hADSC-tumor cells interactions in tumor microenvironment in the design of novel cell therapies.
Collapse
Affiliation(s)
- Giovana Ravizzoni Onzi
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Department of Biophysics and Center of Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Pítia Flores Ledur
- Department of Biophysics and Center of Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Luana Dimer Hainzenreder
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Ana Paula Santin Bertoni
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Andrew Oliveira Silva
- Department of Biophysics and Center of Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Guido Lenz
- Department of Biophysics and Center of Biotechnology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Márcia Rosângela Wink
- Laboratory of Cell Biology, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Gelatin and galactomannan-based scaffolds: Characterization and potential for tissue engineering applications. Carbohydr Polym 2015; 133:8-18. [DOI: 10.1016/j.carbpol.2015.06.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 06/10/2015] [Accepted: 06/12/2015] [Indexed: 11/18/2022]
|
15
|
MA JIANGCHUN, CHENG PENG, HU YI, XUE YIXUE, LIU YUNHUI. Integrin α4 is involved in the regulation of glioma-induced motility of bone marrow mesenchymal stem cells. Oncol Rep 2015; 34:779-86. [DOI: 10.3892/or.2015.4012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/08/2015] [Indexed: 01/14/2023] Open
|
16
|
Souza MC, Silva JD, Pádua TA, Torres ND, Antunes MA, Xisto DG, Abreu TP, Capelozzi VL, Morales MM, Sá Pinheiro AA, Caruso-Neves C, Henriques MG, Rocco PRM. Mesenchymal stromal cell therapy attenuated lung and kidney injury but not brain damage in experimental cerebral malaria. Stem Cell Res Ther 2015; 6:102. [PMID: 25998168 PMCID: PMC4462088 DOI: 10.1186/s13287-015-0093-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 04/01/2015] [Accepted: 05/11/2015] [Indexed: 12/13/2022] Open
Abstract
Introduction Malaria is the most relevant parasitic disease worldwide, and still accounts for 1 million deaths each year. Since current antimalarial drugs are unable to prevent death in severe cases, new therapeutic strategies have been developed. Mesenchymal stromal cells (MSC) confer host resistance against malaria; however, thus far, no study has evaluated the therapeutic effects of MSC therapy on brain and distal organ damage in experimental cerebral malaria. Methods Forty C57BL/6 mice were injected intraperitoneally with 5 × 106Plasmodium berghei-infected erythrocytes or saline. After 24 h, mice received saline or bone marrow (BM)-derived MSC (1x105) intravenously and were housed individually in metabolic cages. After 4 days, lung and kidney morphofunction; cerebrum, spleen, and liver histology; and markers associated with inflammation, fibrogenesis, and epithelial and endothelial cell damage in lung tissue were analyzed. Results In P. berghei-infected mice, BM-MSCs: 1) reduced parasitemia and mortality; 2) increased phagocytic neutrophil content in brain, even though BM-MSCs did not affect the inflammatory process; 3) decreased malaria pigment detection in spleen, liver, and kidney; 4) reduced hepatocyte derangement, with an increased number of Kupffer cells; 5) decreased kidney damage, without effecting significant changes in serum creatinine levels or urinary flow; and 6) reduced neutrophil infiltration, interstitial edema, number of myofibroblasts within interstitial tissue, and collagen deposition in lungs, resulting in decreased lung static elastance. These morphological and functional changes were not associated with changes in levels of tumor necrosis factor-α, keratinocyte-derived chemokine (KC, a mouse analog of interleukin-8), or interferon-γ, which remained increased and similar to those of P. berghei animals treated with saline. BM-MSCs increased hepatocyte growth factor but decreased VEGF in the P. berghei group. Conclusions BM-MSC treatment increased survival and reduced parasitemia and malaria pigment accumulation in spleen, liver, kidney, and lung, but not in brain. The two main organs associated with worse prognosis in malaria, lung and kidney, sustained less histological damage after BM-MSC therapy, with a more pronounced improvement in lung function.
Collapse
Affiliation(s)
- Mariana C Souza
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Johnatas D Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Tatiana A Pádua
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Natália D Torres
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Thiago P Abreu
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Vera L Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, Av. Dr. Arnaldo, 455, Cerqueira César, CEP-01246903, São Paulo, SP, Brazil.
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Ana A Sá Pinheiro
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Celso Caruso-Neves
- Laboratory of Biochemistry and Cellular Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| | - Maria G Henriques
- Laboratory of Applied Pharmacology, Farmanguinhos, Oswaldo Cruz Foundation, Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil. .,National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation (Fiocruz), Av Brasil, 4365, Manguinhos, CEP-21040-900, Rio de Janeiro, RJ, Brazil.
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Av Carlos Chagas Filho, 373 Bloco G, Cidade Universitária, CEP-21941-902, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
17
|
Leite CF, Almeida TR, Lopes CS, Dias da Silva VJ. Multipotent stem cells of the heart-do they have therapeutic promise? Front Physiol 2015; 6:123. [PMID: 26005421 PMCID: PMC4424849 DOI: 10.3389/fphys.2015.00123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 04/06/2015] [Indexed: 01/26/2023] Open
Abstract
The last decade has brought a comprehensive change in our view of cardiac remodeling processes under both physiological and pathological conditions, and cardiac stem cells have become important new players in the general mainframe of cardiac homeostasis. Different types of cardiac stem cells show different capacities for differentiation into the three major cardiac lineages: myocytes, endothelial cells and smooth muscle cells. Physiologically, cardiac stem cells contribute to cardiac homeostasis through continual cellular turnover. Pathologically, these cells exhibit a high level of proliferative activity in an apparent attempt to repair acute cardiac injury, indicating that these cells possess (albeit limited) regenerative potential. In addition to cardiac stem cells, mesenchymal stem cells represent another multipotent cell population in the heart; these cells are located in regions near pericytes and exhibit regenerative, angiogenic, antiapoptotic, and immunosuppressive properties. The discovery of these resident cardiac stem cells was followed by a number of experimental studies in animal models of cardiomyopathies, in which cardiac stem cells were tested as a therapeutic option to overcome the limited transdifferentiating potential of hematopoietic or mesenchymal stem cells derived from bone marrow. The promising results of these studies prompted clinical studies of the role of these cells, which have demonstrated the safety and practicability of cellular therapies for the treatment of heart disease. However, questions remain regarding this new therapeutic approach. Thus, the aim of the present review was to discuss the multitude of different cardiac stem cells that have been identified, their possible functional roles in the cardiac regenerative process, and their potential therapeutic uses in treating cardiac diseases.
Collapse
Affiliation(s)
- Camila F Leite
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Thalles R Almeida
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Carolina S Lopes
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| | - Valdo J Dias da Silva
- Department of Biochemistry, Pharmacology, Physiology and Molecular Biology, Institute for Biological and Natural Sciences, Triângulo Mineiro Federal University Uberaba, Brazil
| |
Collapse
|
18
|
Marx C, Silveira MD, Beyer Nardi N. Adipose-derived stem cells in veterinary medicine: characterization and therapeutic applications. Stem Cells Dev 2015; 24:803-13. [PMID: 25556829 DOI: 10.1089/scd.2014.0407] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells, considered one of the most promising cell types for therapeutic applications due to their capacity to secrete regenerative bioactive molecules, are present in all tissues. Stem cells derived from the adipose tissue have been increasingly used for cell therapy in humans and animals, both as freshly isolated, stromal vascular fraction (SVF) cells, or as cultivated adipose-derived stem cells (ASCs). ASCs have been characterized in different animal species for proliferation, differentiation potential, immunophenotype, gene expression, and potential for tissue engineering. Whereas canine and equine ASCs are well studied, feline cells are still poorly known. Many companies around the world offer ASC therapy for dogs, cats, and horses, although in most countries these activities are not yet controlled by regulatory agencies. This is the first study to review the characterization and clinical use of SVF and ASCs in spontaneously occurring diseases in veterinary patients. Although a relatively large number of studies investigating ASC therapy in induced lesions are available in the literature, a surprisingly small number of reports describe ASC therapy for naturally affected dogs, cats, and horses. A total of seven studies were found with dogs, only two studies in cats, and four in horses. Taken as a whole, the results do not allow a conclusion on the effect of this therapy, due to the generally small number of patients included, diversity of cell populations used, and lack of adequate controls. Further controlled studies are clearly needed to establish the real potential of ASC in veterinary medicine.
Collapse
Affiliation(s)
- Camila Marx
- 1 Laboratory of Stem Cells and Tissue Engineering, Universidade Luterana do Brasil , Canoas, RS, Brazil
| | | | | |
Collapse
|
19
|
Antunes MA, Laffey JG, Pelosi P, Rocco PRM. Mesenchymal stem cell trials for pulmonary diseases. J Cell Biochem 2014; 115:1023-32. [PMID: 24515922 DOI: 10.1002/jcb.24783] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 02/06/2014] [Indexed: 01/12/2023]
Abstract
All adult tissues, including the lung, have some capacity to self-repair or regenerate through the replication and differentiation of stem cells resident within these organs. While lung resident stem cells are an obvious candidate cell therapy for lung diseases, limitations exist regarding our knowledge of the biology of these cells. In contrast, there is considerable interest in the therapeutic potential of exogenous cells, particularly mesenchymal stem/stromal cells (MSCs), for lung diseases. Bone marrow derived-MSCs are the most studied cell therapy for these diseases. Preclinical studies demonstrate promising results using MSCs for diverse lung disorders, including emphysema, bronchopulmonary dysplasia, fibrosis, and acute respiratory distress syndrome. This mini-review will summarize ongoing clinical trials using MSCs in lung diseases, critically examine the data supporting their use for this purpose, and discuss the next steps in the translational pathway for MSC therapy of lung diseases.
Collapse
Affiliation(s)
- Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | |
Collapse
|
20
|
Antunes MA, Abreu SC, Cruz FF, Teixeira AC, Lopes-Pacheco M, Bandeira E, Olsen PC, Diaz BL, Takyia CM, Freitas IPRG, Rocha NN, Capelozzi VL, Xisto DG, Weiss DJ, Morales MM, Rocco PRM. Effects of different mesenchymal stromal cell sources and delivery routes in experimental emphysema. Respir Res 2014; 15:118. [PMID: 25272959 PMCID: PMC4189723 DOI: 10.1186/s12931-014-0118-x] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 09/25/2014] [Indexed: 12/26/2022] Open
Abstract
We sought to assess whether the effects of mesenchymal stromal cells (MSC) on lung inflammation and remodeling in experimental emphysema would differ according to MSC source and administration route. Emphysema was induced in C57BL/6 mice by intratracheal (IT) administration of porcine pancreatic elastase (0.1 UI) weekly for 1 month. After the last elastase instillation, saline or MSCs (1×105), isolated from either mouse bone marrow (BM), adipose tissue (AD) or lung tissue (L), were administered intravenously (IV) or IT. After 1 week, mice were euthanized. Regardless of administration route, MSCs from each source yielded: 1) decreased mean linear intercept, neutrophil infiltration, and cell apoptosis; 2) increased elastic fiber content; 3) reduced alveolar epithelial and endothelial cell damage; and 4) decreased keratinocyte-derived chemokine (KC, a mouse analog of interleukin-8) and transforming growth factor-β levels in lung tissue. In contrast with IV, IT MSC administration further reduced alveolar hyperinflation (BM-MSC) and collagen fiber content (BM-MSC and L-MSC). Intravenous administration of BM- and AD-MSCs reduced the number of M1 macrophages and pulmonary hypertension on echocardiography, while increasing vascular endothelial growth factor. Only BM-MSCs (IV > IT) increased the number of M2 macrophages. In conclusion, different MSC sources and administration routes variably reduced elastase-induced lung damage, but IV administration of BM-MSCs resulted in better cardiovascular function and change of the macrophage phenotype from M1 to M2.
Collapse
Affiliation(s)
- Mariana A Antunes
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| | - Soraia C Abreu
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| | - Fernanda F Cruz
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| | - Ana Clara Teixeira
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| | - Miquéias Lopes-Pacheco
- />Laboratory of Cellular and Molecular Physiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elga Bandeira
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
- />Laboratory of Cellular and Molecular Physiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscilla C Olsen
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| | - Bruno L Diaz
- />Laboratory of Inflammation, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christina M Takyia
- />Laboratory of Cellular Pathology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isalira PRG Freitas
- />Laboratory of Cellular and Molecular Cardiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Vera L Capelozzi
- />Department of Pathology, University of São Paulo, São Paulo, Brazil
| | - Débora G Xisto
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
- />Laboratory of Cellular and Molecular Physiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- />Department of Medicine, University of Vermont, Vermont, USA
| | - Marcelo M Morales
- />Laboratory of Cellular and Molecular Physiology, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia RM Rocco
- />Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Centro de Ciências da Saúde, Federal University of Rio de Janeiro, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão – 21941-902, Rio de Janeiro, RJ Brazil
| |
Collapse
|
21
|
Cunha FF, Martins L, Martin PKM, Stilhano RS, Han SW. A comparison of the reparative and angiogenic properties of mesenchymal stem cells derived from the bone marrow of BALB/c and C57/BL6 mice in a model of limb ischemia. Stem Cell Res Ther 2013; 4:86. [PMID: 23890057 PMCID: PMC3856613 DOI: 10.1186/scrt245] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/23/2013] [Indexed: 11/10/2022] Open
Abstract
Introduction BALB/c mice and C57/BL6 mice have different abilities to recover from ischemia. C57/BL6 mice display increased vessel collateralization and vascular endothelial growth factor expression with a consequent rapid recovery from ischemia compared with BALB/c mice. Mesenchymal stem cells (MSCs) are one of the main cell types that contribute to the recovery from ischemia because, among their biological activities, they produce several proangiogenic paracrine factors and differentiate into endothelial cells. The objective of this study was to evaluate whether the MSCs of these two mouse strains have different inductive capacities for recovering ischemic limbs. Methods MSCs from these two strains were obtained from the bone marrow, purified and characterized before being used for in vivo experiments. Limb ischemia was surgically induced in BALB/c mice, and MSCs were injected on the fifth day. The evolution of limb necrosis was evaluated over the subsequent month. Muscle strength was assessed on the 30th day after the injection, and then the animals were sacrificed to determine the muscle mass and perform histological analyses to detect cellular infiltration, capillary and microvessel densities, fibrosis, necrosis and tissue regeneration. Results The MSCs from both strains promoted high level of angiogenesis similarly, resulting in good recovery from ischemia. However, BALB/c MSCs promoted more muscle regeneration (57%) than C57/BL6 MSCs (44%), which was reflected in the increased muscle strength (0.79 N versus 0.45 N). Conclusion The different genetic background of MSCs from BALB/c mice and C57/BL6 mice was not a relevant factor in promoting angiogenesis of limb ischemia, because both cells showed a similar angiogenic activity. These cells also showed a potential myogenic effect, but the stronger effect promoted by BALB/c MSCs indicates that the different genetic background of MSCs was more relevant in myogenesis than angiogesis.
Collapse
|
22
|
Differences in gene expression and cytokine release profiles highlight the heterogeneity of distinct subsets of adipose tissue-derived stem cells in the subcutaneous and visceral adipose tissue in humans. PLoS One 2013; 8:e57892. [PMID: 23526958 PMCID: PMC3589487 DOI: 10.1371/journal.pone.0057892] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 01/28/2013] [Indexed: 12/13/2022] Open
Abstract
Differences in the inherent properties of adipose tissue-derived stem cells (ASC) may contribute to the biological specificity of the subcutaneous (Sc) and visceral (V) adipose tissue depots. In this study, three distinct subpopulations of ASC, i.e. ASCSVF, ASCBottom, and ASCCeiling, were isolated from Sc and V fat biopsies of non-obese subjects, and their gene expression and functional characteristics were investigated. Genome-wide mRNA expression profiles of ASCSVF, ASCBottom and ASCCeiling from Sc fat were significantly different as compared to their homologous subsets of V-ASCs. Furthermore, ASCSVF, ASCCeiling and ASCBottom from the same fat depot were also distinct from each other. In this respect, both principal component analysis and hierarchical clusters analysis showed that ASCCeiling and ASCSVF shared a similar pattern of closely related genes, which was highly different when compared to that of ASCBottom. However, larger variations in gene expression were found in inter-depot than in intra-depot comparisons. The analysis of connectivity of genes differently expressed in each ASC subset demonstrated that, although there was some overlap, there was also a clear distinction between each Sc-ASC and their corresponding V-ASC subsets, and among ASCSVF, ASCBottom, and ASCCeiling of Sc or V fat depots in regard to networks associated with regulation of cell cycle, cell organization and development, inflammation and metabolic responses. Finally, the release of several cytokines and growth factors in the ASC cultured medium also showed both inter- and intra-depot differences. Thus, ASCCeiling and ASCBottom can be identified as two genetically and functionally heterogeneous ASC populations in addition to the ASCSVF, with ASCBottom showing the highest degree of unmatched gene expression. On the other hand, inter-depot seem to prevail over intra-depot differences in the ASC gene expression assets and network functions, contributing to the high degree of specificity of Sc and V adipose tissue in humans.
Collapse
|
23
|
Comparison of endometrial regenerative cells and bone marrow stromal cells. J Transl Med 2012; 10:207. [PMID: 23038994 PMCID: PMC3504519 DOI: 10.1186/1479-5876-10-207] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/19/2012] [Indexed: 12/17/2022] Open
Abstract
Background Endometrial regenerative cells (ERC) and bone marrow stromal cells (BMSC) are being used in clinical trials. While they have been reported to have similar characteristics, they have not been directly compared. Methods We compared micro RNA (miRNA) and gene expression profiles, soluble cytokine and growth factor levels and ability to inhibit ongoing mixed leukocyte reaction (MLR) of ERC and BMSC each derived from 6 healthy subjects. Results ERC and BMSC miRNA and gene expression profiles were similar, but not identical; more differences were noted in the expression of genes than in miRNAs. Genes overexpressed in ERCs were more likely to be in immune and inflammation pathways and those overexpressed in BMSCs were more likely to be in stem cell and cancer signaling pathways. In addition, the levels of IL-8 and ICAM-1 were greater in ERC supernatants while the levels of HGF, VEGF, IL-6, CXCL12, TGFB1 and TGFB2 were greater in BMSC supernatants. Additionally, ERC demonstrated greater inhibition of the proliferation of mixed leukocyte cultures. Conclusions These results suggest that the in vivo effects of ERC and BMSC may differ. Multiple properties of stromal cells are responsible for their in vivo effectiveness and ERC may be more effective for some of the clinical applications and BMSC for others. Studies in animal models or clinical trials will be required to more fully characterize the differences between ERC and BMSC.
Collapse
|
24
|
Abstract
In this article, development of articular cartilage and endochondral ossification is reviewed, from the perspective of both morphologic aspects of histogenesis and molecular biology, particularly with respect to key signaling molecules and extracellular matrix components most active in cartilage development. The current understanding of the roles of transforming growth factor β and associated signaling molecules, bone morphogenic proteins, and molecules of the Wnt-β catenin system in chondrogenesis are described. Articular cartilage development is a highly conserved complex biological process that is dynamic and robust in nature, which proceeds well without incident or failure in all joints of most young growing individuals.
Collapse
|