1
|
Li C, Wang C, Xie HY, Huang L. Cell-Based Biomaterials for Coronavirus Disease 2019 Prevention and Therapy. Adv Healthc Mater 2023; 12:e2300404. [PMID: 36977465 DOI: 10.1002/adhm.202300404] [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: 02/09/2023] [Revised: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Coronavirus disease 2019 (COVID-19) continues to threaten human health, economic development, and national security. Although many vaccines and drugs have been explored to fight against the major pandemic, their efficacy and safety still need to be improved. Cell-based biomaterials, especially living cells, extracellular vesicles, and cell membranes, offer great potential in preventing and treating COVID-19 owing to their versatility and unique biological functions. In this review, the characteristics and functions of cell-based biomaterials and their biological applications in COVID-19 prevention and therapy are described. First the pathological features of COVID-19 are summarized, providing enlightenment on how to fight against COVID-19. Next, the classification, organization structure, characteristics, and functions of cell-based biomaterials are focused on. Finally, the progress of cell-based biomaterials in overcoming COVID-19 in different aspects, including the prevention of viral infection, inhibition of viral proliferation, anti-inflammation, tissue repair, and alleviation of lymphopenia are comprehensively described. At the end of this review, a look forward to the challenges of this aspect is presented.
Collapse
Affiliation(s)
- Chuyu Li
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Chenguang Wang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Hai-Yan Xie
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| | - Lili Huang
- School of Medical Technology, Beijing Institute of Technology, Beijing, 100081, P. R. China
| |
Collapse
|
2
|
Adipose and Bone Marrow Derived-Mesenchymal Stromal Cells Express Similar Tenogenic Expression Levels when Subjected to Mechanical Uniaxial Stretching In Vitro. Stem Cells Int 2023; 2023:4907230. [PMID: 36756494 PMCID: PMC9902123 DOI: 10.1155/2023/4907230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 05/12/2022] [Accepted: 09/03/2022] [Indexed: 01/31/2023] Open
Abstract
The present study was conducted to determine whether adipose derived mesenchymal stromal cells (AD-MSCs) or bone marrow derived-MSCs (BM-MSCs) would provide superior tenogenic expressions when subjected to cyclical tensile loading. The results for this would indicate the best choice of MSCs source to be used for cell-based tendon repair strategies. Both AD-MSCs and BM-MSCs were obtained from ten adult donors (N = 10) and cultured in vitro. At passaged-2, cells from both groups were subjected to cyclical stretching at 1 Hz and 8% of strain. Cellular morphology, orientation, proliferation rate, protein, and gene expression levels were compared at 0, 24, and 48 hours of stretching. In both groups, mechanical stretching results in similar morphological changes, and the redirection of cell alignment is perpendicular to the direction of stretching. Loading at 8% strain did not significantly increase proliferation rates but caused an increase in total collagen expression and tenogenic gene expression levels. In both groups, these levels demonstrated no significant differences suggesting that in a similar loading environment, both cell types possess similar tenogenic potential. In conclusion, AD-MSCs and BM-MSCs both demonstrate similar tenogenic phenotypic and gene expression levels when subjected to cyclic tensile loading at 1 Hz and 8% strain, thus, suggesting that the use of either cell source may be suitable for tendon repair.
Collapse
|
3
|
Xia Y, Yang R, Hou Y, Wang H, Li Y, Zhu J, Fu C. Application of mesenchymal stem cell-derived exosomes from different sources in intervertebral disc degeneration. Front Bioeng Biotechnol 2022; 10:1019437. [PMID: 36277386 PMCID: PMC9585200 DOI: 10.3389/fbioe.2022.1019437] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is a main cause of lower back pain, leading to psychological and economic burdens to patients. Physical therapy only delays pain in patients but cannot eliminate the cause of IVDD. Surgery is required when the patient cannot tolerate pain or has severe neurological symptoms. Although surgical resection of IVD or decompression of the laminae eliminates the diseased segment, it damages adjacent normal IVD. There is also a risk of re-protrusion after IVD removal. Cell therapy has played a crucial role in the development of regenerative medicine. Cell transplantation promotes regeneration of degenerative tissue. However, owing to the lack of vascular structure in IVD, sufficient nutrients cannot be provided for transplanted mesenchymal stem cells (MSCs). In addition, dead cells release harmful substances that aggravate IVDD. Extracellular vesicles (EVs) have been extensively studied as an emerging therapeutic approach. EVs generated by paracrine MSCs retain the potential of MSCs and serve as carriers to deliver their contents to target cells to regulate target cell activity. Owing to their double-layered membrane structure, EVs have a low immunogenicity and no immune rejection. Therefore, EVs are considered an emerging therapeutic modality in IVDD. However, they are limited by mass production and low loading rates. In this review, the structure of IVD and advantages of EVs are introduced, and the application of MSC-EVs in IVDD is discussed. The current limitations of EVs and future applications are described.
Collapse
Affiliation(s)
- Yuanliang Xia
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Ruohan Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yulin Hou
- Department of Cardiology, Guangyuan Central Hospital, Guangyuan, China
| | - Hengyi Wang
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Yuehong Li
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
| | - Changfeng Fu
- Department of Spine Surgery, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Changfeng Fu,
| |
Collapse
|
4
|
Wu X, Gou H, Zhou O, Qiu H, Liu H, Fu Z, Chen L. Human umbilical cord mesenchymal stem cells combined with pirfenidone upregulates the expression of RGS2 in the pulmonary fibrosis in mice. Respir Res 2022; 23:270. [PMID: 36182915 PMCID: PMC9526322 DOI: 10.1186/s12931-022-02192-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 09/22/2022] [Indexed: 11/10/2022] Open
Abstract
Objective The therapeutic effect of umbilical cord-derived mesenchymal stem cells (hUC-MSCs) in combination with pirfenidone (PFD) on pulmonary fibrosis in mice and its possible mechanism were investigated. Methods C57BL/6 mice were randomly divided into six groups: control group, model group, P10 group, P30 group, P100 group, and P300 group. Modeled by tracheal intubation with 3 mg/kg bleomycin drip, each dose of PFD was administered daily by gavage from day 7 onwards. The mice were observed continuously for 21 days and survival was recorded. Lung tissues were collected on day 21, and hematoxylin–eosin (HE) and Masson staining were performed to assess morphological changes and collagen deposition in the lungs. Collagen content was measured by the Sircol method, and fibrosis marker levels were detected by PCR and Western blot. Another batch of C57BL/6 mice was then randomly divided into five groups: hUC-MSC control group, model group, P100 group, hUC-MSC treatment group, and hUC-MSCs + P30 group. On day 7, 5 × 105 hUC-MSCs were injected into the tail vein, the mice were administered PFD gavage daily from day 7 onwards, and their survival was recorded. Lung tissues were collected on day 21 to detect pathological changes, the collagen content, and the expression of regulator of G protein signaling 2 (RGS2). Pulmonary myofibroblasts (MFBs) were divided into an MFB group and an MFB + hUC-MSCs group; different doses of PFD were administered to each group, and the levels of RGS2, intracellular Ca2+, and fibrosis markers were recorded for each group. Results Compared with other PFD group doses, the P100 group had significantly improved mouse survival and lung pathology and significantly reduced collagen and fibrosis marker levels (p < 0.05). The hUC-MSCs + P30 group had significantly improved mouse survival and lung pathology, significantly reduced collagen content and fibrosis marker levels (p < 0.05), and the efficacy was better than that of the P100 and hUC-MSCs groups (p < 0.05). RGS2 expression was significantly higher in the MSCs + P30 group compared with the P100 and hUC-MSCs groups (p < 0.05). PFD increased RGS2 expression in MFBs (p < 0.05) in a dose-dependent manner. Compared with PFD and hUC-MSCs treatment alone, combination of hUC-MSCs and PFD increased RGS2 protein levels, significantly decreased intracellular Ca2+ concentration, and significantly reduced fibrosis markers. Conclusion The findings suggest that hUC-MSCs combined with low-dose PFD have a therapeutic effect better than that of the two treatments used separately. Its effect on attenuating bleomycin-induced pulmonary fibrosis in mice is related to the increase of RGS2. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02192-6.
Collapse
Affiliation(s)
- Xian Wu
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Hao Gou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
| | - Ou Zhou
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Huijun Qiu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Hanmin Liu
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China.,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Zhou Fu
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China. .,Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing, 400015, China. .,Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing, 400015, China.
| | - Lina Chen
- Division of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China. .,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, Sichuan, China. .,NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, 610065, Sichuan, China.
| |
Collapse
|
5
|
Chiarotto GB, Cartarozzi LP, Perez M, Tomiyama ALMR, de Castro MV, Duarte ASS, Luzo ÂCM, Oliveira ALRD. Delayed onset, immunomodulation, and lifespan improvement of SOD1 G93A mice after intravenous injection of human mesenchymal stem cells derived from adipose tissue. Brain Res Bull 2022; 186:153-164. [PMID: 35718222 DOI: 10.1016/j.brainresbull.2022.06.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/07/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease characterized by the selective and progressive loss of motor neurons from the spinal cord, brain stem, and motor cortex. Although the hallmark of ALS is motor neuron degeneration, astrocytes, microglia, and T cells actively participate. Pharmacological treatment with riluzole has little effect on the lifespan of the patient. Thus, the development of new therapeutic strategies is of utmost importance. The objective of this study was to verify whether human mesenchymal stem cells (hMSCs) from adipose tissue have therapeutic potential in SOD1G93A transgenic mice. The treatment was carried out in the asymptomatic phase of the disease (10th week) by a single systemic application of ad-hMSCs (1 ×105 cells). The animals were sacrificed at the 14th week (the initial stage of symptoms) or the end-stage (ES) of the disease. The lumbar spinal cords were dissected and processed for Nissl staining (neuronal survival), immunohistochemistry (gliosis and synaptic preservation), and gene transcript expression (qRT-PCR). Behavioral analyses considering the onset of disease and its progression, neurological score, body weight, and motor control (rotarod test) started on the 10th week and were performed every three days until the ES of the disease. The results revealed that treatment with ad-hMSCs promoted greater neuronal survival (44%) than vehicle treatment. However, no effect was seen at the ES of the disease. Better structural preservation of the ventral horn in animals treated with ad-hMSCs was observed, together with decreased gliosis and greater synapse protection. In line with this, we found that the transcript levels of Hgf1 were upregulated in ad-hMSCs-treated mice. These results corroborate the behavioral data showing that ad-hMSCs had delayed motor deficits and reduced weight loss compared to vehicle animals. Additionally, cell therapy delayed the course of the disease and significantly improved survival by 20 days. Overall, our results indicate that treatment with ad-hMSCs has beneficial effects, enhancing neuronal survival and promoting a less degenerative neuronal microenvironment. Thus, this may be a potential therapy to improve the quality of life and to extend the lifespan of ALS patients.
Collapse
Affiliation(s)
- Gabriela Bortolança Chiarotto
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil
| | - Luciana Politti Cartarozzi
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil
| | - Matheus Perez
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, 14040-907 Ribeirão Preto, SP, Brazil
| | - Ana Laura Midori Rossi Tomiyama
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil
| | - Mateus Vidigal de Castro
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil
| | - Adriana S S Duarte
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Ângela Cristina Malheiros Luzo
- Hematology and Hemotherapy Center, University of Campinas/Hemocentro-Unicamp, Instituto Nacional de Ciência e Tecnologia do Sangue, Campinas, São Paulo, Brazil
| | - Alexandre Leite Rodrigues de Oliveira
- Laboratory of Nerve Regeneration, University of Campinas - UNICAMP, Cidade Universitaria "Zeferino Vaz", Rua Monteiro Lobato, 255, Campinas, 13083-862, SP, Brazil.
| |
Collapse
|
6
|
Fan M, Shi H, Yao H, Wang W, Zhang Y, Jiang C, Lin R. Glutamate regulates gliosis of BMSCs to promote ENS regeneration through α-KG and H3K9/H3K27 demethylation. Stem Cell Res Ther 2022; 13:255. [PMID: 35715822 PMCID: PMC9205030 DOI: 10.1186/s13287-022-02936-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/19/2022] [Indexed: 11/18/2022] Open
Abstract
Background There is a lack of effective therapies for enteric nervous system (ENS) injury. Our previous study showed that transplanted bone marrow-derived mesenchymal stem cells (BMSCs) play a “glia-like cells” role in initiating ENS regeneration in denervated mice. Cellular energy metabolism is an important factor in maintaining the biological characteristics of stem cells. However, how cellular energy metabolism regulates the fate of BMSCs in the ENS-injured microenvironment is unclear. Methods The biological characteristics, energy metabolism, and histone methylation levels of BMSCs following ENS injury were determined. Then, glutamate dehydrogenase 1 (Glud1) which catalyzes the oxidative deamination of glutamate to α-KG was overexpressed (OE) in BMSCs. Further, OE-Glud1 BMSCs were targeted–transplanted into the ENS injury site of denervated mice to determine their effects on ENS regeneration. Results In vitro, in the ENS-injured high-glutamate microenvironment, the ratio of α-ketoglutarate (α-KG) to succinate (P < 0.05), the histone demethylation level (P < 0.05), the protein expression of glial cell markers (P < 0.05), and the gene expression of Glud1 (P < 0.05) were significantly increased. And the binding of H3K9me3 to the GFAP, S100B, and GDNF promoter was enhanced (P < 0.05). Moreover, α-KG treatment increased the monomethylation and decreased the trimethylation on H3K9 (P < 0.01) and H3K27 (P < 0.05) in BMSCs and significantly upregulated the protein expression of glial cell markers (P < 0.01), which was reversed by the α-KG competitive inhibitor D-2-hydroxyglutarate (P < 0.05). Besides, overexpression of Glud1 in BMSCs exhibited increases in monomethylation and decreases in trimethylation on H3K9 (P < 0.05) and H3K27 (P < 0.05), and upregulated protein expression of glial cell markers (P < 0.01). In vivo, BMSCs overexpressing Glud1 had a strong promotion effect on ENS regeneration in denervated mice through H3K9/H3K27 demethylation (P < 0.05), and upregulating the expression of glial cell protein (P < 0.05). Conclusions BMSCs overexpressing Glud1 promote the expression of glial cell markers and ENS remodeling in denervated mice through regulating intracellular α-KG and H3K9/H3K27 demethylation. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02936-7.
Collapse
Affiliation(s)
- Mengke Fan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huiying Shi
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hailing Yao
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weijun Wang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yurui Zhang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chen Jiang
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Rong Lin
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
7
|
Salame N, Bikorimana JP, El-Hachem N, Saad W, Kurdi M, Zhao J, Eliopoulos N, Shammaa R, Rafei M. UM171A-induced ROS promote antigen cross-presentation of immunogenic peptides by bone marrow-derived mesenchymal stromal cells. Stem Cell Res Ther 2022; 13:16. [PMID: 35012668 PMCID: PMC8751335 DOI: 10.1186/s13287-021-02693-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/24/2021] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have been extensively used in the clinic due to their exquisite tissue repair capacity. However, they also hold promise in the field of cellular vaccination as they can behave as conditional antigen presenting cells in response to interferon (IFN)-gamma treatment under a specific treatment regimen. This suggests that the immune function of MSCs can be pharmacologically modulated. Given the capacity of the agonist pyrimido-indole derivative UM171a to trigger the expression of various antigen presentation-related genes in human hematopoietic progenitor cells, we explored the potential use of UM171a as a means to pharmacologically instill and/or promote antigen presentation by MSCs. Methods Besides completing a series of flow-cytometry-based phenotypic analyses, several functional antigen presentation assays were conducted using the SIINFEKL-specific T-cell clone B3Z. Anti-oxidants and electron transport chain inhibitors were also used to decipher UM171a’s mode of action in MSCs. Finally, the potency of UM171a-treated MSCs was evaluated in the context of therapeutic vaccination using immunocompetent C57BL/6 mice with pre-established syngeneic EG.7T-cell lymphoma. Results Treatment of MSCs with UM171a triggered potent increase in H2-Kb cell surface levels along with the acquisition of antigen cross-presentation abilities. Mechanistically, such effects occurred in response to UM171a-mediated production of mitochondrial-derived reactive oxygen species as their neutralization using anti-oxidants or Antimycin-A mitigated MSCs’ ability to cross-present antigens. Processing and presentation of the immunogenic ovalbumin-derived SIINFEKL peptide was caused by de novo expression of the Psmb8 gene in response to UM171a-triggered oxidative stress. When evaluated for their anti-tumoral properties in the context of therapeutic vaccination, UM171a-treated MSC administration to immunocompetent mice with pre-established T-cell lymphoma controlled tumor growth resulting in 40% survival without the need of additional supportive therapy and/or standard-of-care. Conclusions Altogether, our findings reveal a new immune-related function for UM171a and clearly allude to a direct link between UM171a-mediated ROS induction and antigen cross-presentation by MSCs. The fact that UM171a treatment modulates MSCs to become antigen-presenting cells without the use of IFN-gamma opens-up a new line of investigation to search for additional agents capable of converting immune-suppressive MSCs to a cellular tool easily adaptable to vaccination. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02693-z.
Collapse
Affiliation(s)
- Natasha Salame
- Department of Biomedical Sciences, Université de Montréal, Montreal, QC, Canada
| | - Jean-Pierre Bikorimana
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada
| | - Nehme El-Hachem
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada.,Pediatric Hematology-Oncology Division, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, QC, Canada
| | - Wael Saad
- Laboratory of Experimental and Clinical Pharmacology, Department of Chemistry and Biochemistry, Faculty of Sciences, Lebanese University, Hadat, Lebanon
| | - Mazen Kurdi
- Laboratory of Experimental and Clinical Pharmacology, Department of Chemistry and Biochemistry, Faculty of Sciences, Lebanese University, Hadat, Lebanon
| | - Jing Zhao
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Nicoletta Eliopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada.,Department of Surgery, McGill University, Montreal, QC, Canada
| | - Riam Shammaa
- Department of Family and Community Medicine, University of Toronto, Toronto, ON, Canada.,Canadian Centers for Regenerative Therapy, Toronto, ON, Canada.,IntelliStem Technologies Inc., Toronto, ON, Canada
| | - Moutih Rafei
- Department of Microbiology, Infectious Diseases and Immunology, Université de Montréal, Montreal, QC, Canada. .,Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada. .,Molecular Biology Program, Université de Montréal, Montreal, QC, Canada. .,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Static Magnetic Fields Enhance the Chondrogenesis of Mandibular Bone Marrow Mesenchymal Stem Cells in Coculture Systems. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9962861. [PMID: 34873576 PMCID: PMC8643226 DOI: 10.1155/2021/9962861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 10/20/2021] [Accepted: 11/12/2021] [Indexed: 11/18/2022]
Abstract
Objectives Combining the advantages of static magnetic fields (SMF) and coculture systems, we investigated the effect of moderate-intensity SMF on the chondrogenesis and proliferation of mandibular bone marrow mesenchymal stem cells (MBMSCs) in the MBMSC/mandibular condylar chondrocyte (MCC) coculture system. The main aim of the present study was to provide an experimental basis for obtaining better cartilage tissue engineering seed cells for the effective repair of condylar cartilage defects in clinical practice. Methods MBMSCs and MCCs were isolated from SD (Sprague Dawley) rats. Flow cytometry, three-lineage differentiation, colony-forming assays, immunocytochemistry, and toluidine blue staining were used for the identification of MBMSCs and MCCs. MBMSCs and MCCs were seeded into the lower and upper Transwell chambers, respectively, at a ratio of 1 : 2, and exposed to a 280 mT SMF. MBMSCs were harvested after 3, 7, or 14 days for analysis. CCK-8 was used to detect cell proliferation, Alcian blue staining was utilized to evaluate glycosaminoglycan (GAG), and western blotting and real-time quantitative polymerase chain reaction (RT-qPCR) detected protein and gene expression levels of SOX9, Col2A1 (Collagen Type II Alpha 1), and Aggrecan (ACAN). Results The proliferation of MBMSCs was significantly enhanced in the experimental group with MBMSCs cocultured with MCCs under SMF stimulation relative to controls (P < 0.05). GAG content was increased, and SOX9, Col2A1, and ACAN were also increased at the mRNA and protein levels (P < 0.05). Conclusions Moderate-intensity SMF improved the chondrogenesis and proliferation of MBMSCs in the coculture system, and it might be a promising approach to repair condylar cartilage defects in the clinical setting.
Collapse
|
9
|
do Prado-Lima PAS, Costa-Ferro ZSM, Souza BSDF, da Cruz IBM, Lab B. Is there a place for cellular therapy in depression? World J Psychiatry 2021; 11:553-567. [PMID: 34631460 PMCID: PMC8474995 DOI: 10.5498/wjp.v11.i9.553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/05/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Although efforts have been made to improve the pharmacological treatment of depression, approximately one-third of patients with depression do not respond to conventional therapy using antidepressants. Other potential non-pharmacological therapies have been studied in the last years, including the use of mesenchymal stem cell therapies to treat depression. These therapies are reviewed here since it is clinically relevant to develop innovative therapeutics to treat psychiatric patients. Experimental data corroborate that mesenchymal stem cell therapy could be considered a potential treatment for depression based on its anti-inflammatory and neurotrophic properties. However, some clinical trials involving treatment of depression with stem cells are in progress, but with no published results. These studies and other future clinical investigations will be crucial to define how much mesenchymal stem cells can effectively be used in psychiatric clinics as a strategy for supporting depression treatment.
Collapse
Affiliation(s)
- Pedro Antônio Schmidt do Prado-Lima
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Rio Grande do Sul, Brazil
| | - Zaquer Suzana Munhoz Costa-Ferro
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Rio Grande do Sul, Brazil
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, Bahia, Brazil
| | - Bruno Solano de Freitas Souza
- Center for Biotechnology and Cell Therapy, São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
- D’Or Institute for Research and Education (IDOR), Salvador 41253-190, Bahia, Brazil
- Laboratory of Tissue Engineering and Immunopharmacology, Gonçalo Moniz Institute, Fiocruz, Salvador 40296-710, Bahia, Brazil
| | | | - Biogenomics Lab
- Health Sciences Center, Federal University of Santa Maria, Santa Maria 97105900, RS, Brazil
| |
Collapse
|
10
|
Herman S, Fishel I, Offen D. Intranasal delivery of mesenchymal stem cells-derived extracellular vesicles for the treatment of neurological diseases. Stem Cells 2021; 39:1589-1600. [PMID: 34520591 DOI: 10.1002/stem.3456] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 09/02/2021] [Indexed: 12/22/2022]
Abstract
Neurological disorders are diseases of the central nervous system (CNS), characterized by a progressive degeneration of cells and deficiencies in neural functions. Mesenchymal stem cells (MSCs) are a promising therapy for diseases and disorders of the CNS. Increasing evidence suggests that their beneficial abilities can be attributed to their paracrine secretion of extracellular vesicles (EVs). Administration of EVs that contain a mixture of proteins, lipids, and nucleic acids, resembling the secretome of MSCs, has been shown to mimic most of the effects of the parental cells. Moreover, the small size and safety profile of EVs provide a number of advantages over cell transplantation. Intranasal (IN) administration of EVs has been established as an effective and reliable way to bypass the blood-brain barrier (BBB) and deliver drugs to the CNS. In addition to pharmacological drugs, EVs can be loaded with a diverse range of cargo designed to modulate gene expression and protein functions in recipient cells, and lead to immunomodulation, neurogenesis, neuroprotection, and degradation of protein aggregates. In this review, we will explore the proposed physiological pathways by which EVs migrate through the nasal route to the CNS where they can actively target a region of injury or inflammation and exert their therapeutic effects. We will summarize the functional outcomes observed in animal models of neurological diseases following IN treatment with MSC-derived EVs. We will also examine key mechanisms that have been suggested to mediate the beneficial effects of EV-based therapy.
Collapse
Affiliation(s)
- Shay Herman
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Idan Fishel
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Daniel Offen
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Felsenstein Medical Research Center, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Xiao Y, Zhu Q, Liu X, Jiang M, Hao H, Zhu H, Cowan PJ, He X, Liu Q, Zhou S, Liu Z. High-fat diet selectively decreases bone marrow lin - /CD117 + cell population in aging mice through increased ROS production. J Tissue Eng Regen Med 2020; 14:884-892. [PMID: 32337800 DOI: 10.1002/term.3047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 04/03/2020] [Accepted: 04/15/2020] [Indexed: 12/18/2022]
Abstract
Bone marrow (BM) stem cells (BMSCs) are an important source for cell therapy. The outcome of cell therapy could be ultimately associated with the number and function of donor BMSCs. The present study was to evaluate the effect of long-term high-fat diet (HFD) on the population of BMSCs and the role of reactive oxygen species (ROS) in aging mice. Forty-week-old male C57BL/6 mice were fed with HFD for 3 months with regular diet as control. Experiments were repeated when ROS production was reduced in mice treated with N-acetylcysteine (NAC) or using mice overexpressing antioxidant enzyme network (AON) of superoxide dismutase (SOD)1, SOD3, and glutathione peroxidase. BM and blood cells were analyzed with flowcytometry for lineage negative (lin- ) and Sca-1+ , or lin- /CD117+ , or lin- /CD133+ cells. Lin- /CD117+ cell population was significantly decreased with increased intracellular ROS and apoptosis and decreased proliferation in BM, not in blood, in HFD-treated mice without change for Sca-1+ or CD133+ cell populations in BM or blood. NAC treatment or AON overexpression effectively prevented HFD-induced intracellular ROS production and reduction of BM lin- /CD117+ population. These data suggested that long-term HFD selectively decreased BM lin- /CD117+ cell population in aging mice through increased ROS production.
Collapse
Affiliation(s)
- Yichao Xiao
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Qingyi Zhu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China.,Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Xuanyou Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Meng Jiang
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Hua Zhu
- Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH, USA
| | - Peter J Cowan
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia.,Immunology Research Centre, St. Vincent's Hospital, Melbourne, Victoria, Australia
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shenghua Zhou
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
12
|
Human induced pluripotent stem cell-derived mesenchymal stem cells promote healing via TNF-α-stimulated gene-6 in inflammatory bowel disease models. Cell Death Dis 2019; 10:718. [PMID: 31558705 PMCID: PMC6763610 DOI: 10.1038/s41419-019-1957-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/30/2019] [Accepted: 09/02/2019] [Indexed: 12/12/2022]
Abstract
Therapeutic applications of tissue-derived mesenchymal stem cells (MSCs) are hindered by their limited expansion ability and variation across donors. Human induced pluripotent stem cell (iPSC)-derived MSCs show greater expandability and therefore offer potential for use in tissue repair therapies. Here we explored the regenerative effects of iPSC-MSCs and the mechanisms by which iPSC-MSCs promote mucosal healing via tumor necrosis factor-α-stimulated gene 6 (TSG-6) in mouse models of inflammatory bowel disease (IBD). Human iPSCs were induced to differentiate into MSCs following a clinically compliant protocol. The iPSC-MSC treatment promoted mucosal healing in colitic mice, accompanied by increased epithelial cell proliferation, CD44-positive cells, and Lgr5-positive cells. TSG-6 knockdown in iPSC-MSCs or blocking of hyaluronan–CD44 interactions by PEP-1 abrogated the therapeutic effects of iPSC-MSCs, whereas use of recombinant TSG-6 showed therapeutic effects similar to those of iPSC-MSCs. A mouse or patient-derived organoid culture system was developed. Organoids co-cultured with iPSC-MSCs showed increased epithelial cell proliferation, CD44-positive cells, and Lgr5-positive cells, which was abolished by TSG-6 knockdown. TSG-6-induced promoting effects in organoids were dependent on Akt activation and abrogated by the anti-CD44 antibody or MK2206. In conclusion, iPSC-MSCs promoted epithelial cell proliferation to accelerate mucosal healing in a murine colitis model via TSG-6 through hyaluronan–CD44 interactions in an Akt-dependent manner, demonstrating a patient-specific “off-the-shelf” format for IBD treatment.
Collapse
|
13
|
Zhang L, Wang Y, Zhou N, Feng Y, Yang X. Cyclic tensile stress promotes osteogenic differentiation of adipose stem cells via ERK and p38 pathways. Stem Cell Res 2019; 37:101433. [PMID: 31005788 DOI: 10.1016/j.scr.2019.101433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/03/2019] [Accepted: 04/06/2019] [Indexed: 02/05/2023] Open
Abstract
The present study aimed to elucidate whether extracellular signal-regulated kinases 1/2 (ERK1/2) and p38 mitogen-activated protein kinases pathways participate in the transduction of mechanical stretch exerted on adipose stem cells (ASCs) into intracellular osteogenic signals, and if so whether both pathways have time-dependent feature. Rat ASCs were cultured in osteogenic medium for 72 h and assigned into three sets, namely ERK1/2 inhibitor treated set, p38 inhibitor treated set, and the control set. After inhibitor treatment, all cells were subjected to cyclic stretch(2000 με, 1 Hz) on a four-point bending mechanical loading device. Protein and mRNA samples were acquired at six time points: 0, 15 min, 30 min, 1 h, 2 h and 6 h. Western blot showed phosphorylation level of ERK1/2 was elevated by cyclic tensile stress at all time points, while p38 at 15 min, 30 min and 1 h, and the elevation can be completely blocked by corresponding inhibitors. The treatment by ERK1/2 inhibitor was shown to antagonize the up-regulation of osteogenic genes bone morphogenetic protein 2 (BMP-2) and runt-related transcription factor 2 (Runx2) by mechanical stretch at 15 min and 6 h, whereas p38 inhibitor took effect at 15 min only. The results suggested both ERK and p38 could be positive mediators of stretch-induced osteogenic differentiation of ASCs, and ERK stimulate the stretch-induced osteogenic differentiation at both early and late stages while p38 responds to mechanical stretch in a more rapid fashion.
Collapse
Affiliation(s)
- Liang Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yingkai Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Nan Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuzhang Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xingmei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Yu Z, Xu N, Zhang N, Xiong Y, Wang Z, Liang S, Zhao D, Huang F, Zhang C. Repair of Peripheral Nerve Sensory Impairments via the Transplantation of Bone Marrow Neural Tissue-Committed Stem Cell-Derived Sensory Neurons. Cell Mol Neurobiol 2019; 39:341-353. [PMID: 30684112 DOI: 10.1007/s10571-019-00650-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/04/2019] [Indexed: 01/20/2023]
Abstract
The present study aimed to investigate the efficacy of transplantation of bone marrow neural tissue-committed stem cell-derived sensory neuron-like cells for the repair of peripheral nerve sensory impairments in rats. Bone marrow was isolated and cultured to obtain the neural tissue-committed stem cells (NTCSCs), and the differentiation of these cells into sensory neuron-like cells was induced. Bone marrow mesenchymal stem cells (BMSCs), bone marrow NTCSCs, and bone marrow NTCSC-derived sensory neurons (NTCSC-SNs) were transplanted by microinjection into the L4 and L5 dorsal root ganglions (DRGs) in an animal model of sensory defect. On the 2nd, 4th, 8th, and 12th week after the transplantation, the effects of the three types of stem cells on the repair of the sensory functional defect were analyzed via behavioral observation, sensory function evaluation, electrophysiological examination of the sciatic nerve, and morphological observation of the DRGs. The results revealed that the transplanted BMSCs, NTCSCs, and NTCSC-SNs were all able to repair the sensory nerves. In addition, the effect of the NTCSC-SNs was significantly better than that of the other two types of stem cells. The general posture and gait of the animals in the sensory defect model exhibited evident improvement over time. Plantar temperature sensitivity and pain sensitivity gradually recovered, and the sensation latency was reduced, with faster sensory nerve conduction velocity. Transplantation of NTCSC-SNs can improve the repair of peripheral nerve sensory defects in rats.
Collapse
Affiliation(s)
- Zhenhai Yu
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
- Department of Human Anatomy, College of Basic Medical Sciences, Second Military Medical University, Shanghai, 200433, People's Republic of China
| | - Ning Xu
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, People's Republic of China
| | - Naili Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Yanlian Xiong
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Zhiqiang Wang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Shaohua Liang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Dongmei Zhao
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Fei Huang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| | - Chuansen Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China.
- Department of Human Anatomy, College of Basic Medical Sciences, Second Military Medical University, Shanghai, 200433, People's Republic of China.
| |
Collapse
|
15
|
Mu J, Bakreen A, Juntunen M, Korhonen P, Oinonen E, Cui L, Myllyniemi M, Zhao S, Miettinen S, Jolkkonen J. Combined Adipose Tissue-Derived Mesenchymal Stem Cell Therapy and Rehabilitation in Experimental Stroke. Front Neurol 2019; 10:235. [PMID: 30972000 PMCID: PMC6443824 DOI: 10.3389/fneur.2019.00235] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/22/2019] [Indexed: 01/12/2023] Open
Abstract
Background/Objective: Stroke is a leading global cause of adult disability. As the population ages as well as suffers co-morbidities, it is expected that the stroke burden will increase further. There are no established safe and effective restorative treatments to facilitate a good functional outcome in stroke patients. Cell-based therapies, which have a wide therapeutic window, might benefit a large percentage of patients, especially if combined with different restorative strategies. In this study, we tested whether the therapeutic effect of human adipose tissue-derived mesenchymal stem cells (ADMSCs) could be further enhanced by rehabilitation in an experimental model of stroke. Methods: Focal cerebral ischemia was induced in adult male Sprague Dawley rats by permanently occluding the distal middle cerebral artery (MCAO). After the intravenous infusion of vehicle (n = 46) or ADMSCs (2 × 106) either at 2 (n = 37) or 7 (n = 7) days after the operation, half of the animals were housed in an enriched environment mimicking rehabilitation. Subsequently, their behavioral recovery was assessed by a neurological score, and performance in the cylinder and sticky label tests during a 42-day behavioral follow-up. At the end of the follow-up, rats were perfused for histology to assess the extent of angiogenesis (RECA-1), gliosis (GFAP), and glial scar formation. Results: No adverse effects were observed during the follow-up. Combined ADMSC therapy and rehabilitation improved forelimb use in the cylinder test in comparison to MCAO controls on post-operative days 21 and 42 (P < 0.01). In the sticky label test, ADMSCs and rehabilitation alone or together, significantly decreased the removal time as compared to MCAO controls on post-operative days 21 and 42. An early initiation of combined therapy seemed to be more effective. Infarct size, measured by MRI on post-operative days 1 and 43, did not differ between the experimental groups. Stereological counting revealed an ischemia-induced increase both in the density of blood vessels and the numbers of glial cells in the perilesional cortex, but there were no differences among MCAO groups. Glial scar volume was also similar in MCAO groups. Conclusion: Early delivery of ADMSCs and combined rehabilitation enhanced behavioral recovery in an experimental stroke model. The mechanisms underlying these treatment effects remain unknown.
Collapse
Affiliation(s)
- Jingwei Mu
- Department of Neurology, The People's Hospital of China Medical University, Shenyang, China.,Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - Miia Juntunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Paula Korhonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ella Oinonen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Lili Cui
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Myllyniemi
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Shanshan Zhao
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Susanna Miettinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Jukka Jolkkonen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland.,A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
16
|
Kwak KA, Lee SP, Yang JY, Park YS. Current Perspectives regarding Stem Cell-Based Therapy for Alzheimer's Disease. Stem Cells Int 2018; 2018:6392986. [PMID: 29686714 PMCID: PMC5852851 DOI: 10.1155/2018/6392986] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/15/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder featuring memory loss and cognitive impairment, is caused by synaptic failure and the excessive accumulation of misfolded proteins. Many unsuccessful attempts have been made to develop new small molecules or antibodies to intervene in the disease's pathogenesis. Stem cell-based therapies cast a new hope for AD treatment as a replacement or regeneration strategy. The results from recent preclinical studies regarding stem cell-based therapies are promising. Human clinical trials are now underway. However, a number of questions remain to be answered prior to safe and effective clinical translation. This review explores the pathophysiology of AD and summarizes the relevant stem cell research according to cell type. We also briefly summarize related clinical trials. Finally, future perspectives are discussed with regard to their clinical applications.
Collapse
Affiliation(s)
- Kyeong-Ah Kwak
- Department of Oral Anatomy, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Seung-Pyo Lee
- Department of Oral Anatomy, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jin-Young Yang
- Department of Dental Hygiene, Daejeon Institute of Science and Technology, Daejeon, Republic of Korea
| | - Young-Seok Park
- Department of Oral Anatomy, Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
17
|
Menéndez-Menéndez Y, Otero-Hernández J, Vega JA, Pérez-Basterrechea M, Pérez-López S, Álvarez-Viejo M, Ferrero-Gutiérrez A. The role of bone marrow mononuclear cell-conditioned medium in the proliferation and migration of human dermal fibroblasts. Cell Mol Biol Lett 2017; 22:29. [PMID: 29270201 PMCID: PMC5735620 DOI: 10.1186/s11658-017-0055-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/10/2017] [Indexed: 12/14/2022] Open
Abstract
Background Several recent studies have demonstrated the great potential of bone marrow cells in regenerative medicine, not only for their ability to differentiate to match a damaged cell type, but also because they synthesize and release various growth factors and cytokines. We examined the effect of bone marrow cell-conditioned medium in the healing process, especially in terms of fibroblast proliferation and migration. Methods These in vitro studies consisted of co-culture (without direct contact) of dermal fibroblasts with mononuclear bone marrow cells and the use of conditioned medium obtained from these cultures in a scratch wound model. Results Mononuclear cells were found to increase the proliferation of fibroblasts, and the conditioned medium showed a stimulatory effect on the migration of fibroblasts. Conclusion When considered together with the observed increase in growth factor levels in conditioned medium, it appears that these cells act through a paracrine mechanism.
Collapse
Affiliation(s)
- Yolanda Menéndez-Menéndez
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jesús Otero-Hernández
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jose Antonio Vega
- Departamento de Morfología y Biología Celular, Universidad de Oviedo, Oviedo, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Temuco, Chile
| | - Marcos Pérez-Basterrechea
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Silvia Pérez-López
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - María Álvarez-Viejo
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Amaia Ferrero-Gutiérrez
- Unidad de Coordinación de Trasplantes, Terapia Celular y Medicina Regenerativa, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
18
|
Wang X, Gao JL, Zhao MM, Zhu HX, Tian YX, Li R, Jiang XH, Yu L, Tian JR, Cui JZ. Therapeutic effects of conditioned medium from bone marrow-derived mesenchymal stem cells on epithelial-mesenchymal transition in A549 cells. Int J Mol Med 2017; 41:659-668. [PMID: 29207055 PMCID: PMC5752235 DOI: 10.3892/ijmm.2017.3284] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/04/2017] [Indexed: 01/07/2023] Open
Abstract
Pulmonary fibrosis (PF) is a chronic lung disease. The transforming growth factor-β1 (TGF-β1)/Smad3 signaling pathway plays an important role in the pathogenesis of pulmonary fibrosis. Bone marrow-derived mesenchymal stem cells (BMSCs) have been shown to be a modulator of the molecular aspects of the fibrosis pathway. However, it is still unknown as to whether the conditioned medium from BMSCs (BMSCs-CM) inhibits the epithelial-mesenchymal transition (EMT) process. This study confirmed the hypothesis that BMSCs-CM exerts an anti-fibrotic effect on human type II alveolar epithelial cells (A549) by suppressing the phosphorylation of Smad3. We used the A549 cells in vitro to detect morphological evidence of EMT by phase-contrast microscopy. These cells were randomly divided into 4 groups as follows: the control group, the TGF-β1 group, the SIS3 (specific inhibitor of Smad3) group and the BMSCs-CM group. The immunofluorescence method was used to determined the location of E-cadherin (E-calcium mucins; E-cad), α-smooth muscle actin (α-SMA) and p-Smad3. The expression levels of E-cad, CK8, α-SMA, vimentin, p-Smad3, Snail1, collagen I (COLI) and collagen III (COLIII) were detected by western blot analysis. Following exposure to TGF-β1, the A549 cells displayed a spindle-shaped fibroblast-like morphology. In accordance with these morphological changes, the expression levels of E-cad and CK8 were downregulated, while the expression levels of α-SMA and vimentin were upregulated. Along with this process, the expression levels of p-Smad3, Snail1, COLI and COLIII were increased. However, the cells in the BMSCs-CM group and SIS3 group exhibited a decrease in the levels of α-SMA and vimentin (which had been upregulated by TGF-β1), and an increase in the levels of E-cad and CK8 expression (which had been downregulated by TGF-β1). On the whole, these results indicated that BMSCs-CM suppressed the EMT which might be associated with TGF-β1/Smad3. This study provides the theoretical basis for the research of the mechanisms responsible for pulmonary disease.
Collapse
Affiliation(s)
- Xin Wang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Jun-Ling Gao
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Man-Man Zhao
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Hui-Xing Zhu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Yan-Xia Tian
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Ran Li
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Xiao-Hua Jiang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Lei Yu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Jing-Rui Tian
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Jian-Zhong Cui
- Department of Neurosurgery, Tangshan Workers' Hospital, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
19
|
Crowley MG, Tajiri N. Exogenous stem cells pioneer a biobridge to the advantage of host brain cells following stroke: New insights for clinical applications. Brain Circ 2017; 3:130-134. [PMID: 30276314 PMCID: PMC6057688 DOI: 10.4103/bc.bc_17_17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 01/01/2023] Open
Abstract
Stroke continues to maintain its status as one of the top causes of mortality within the United States. Currently, the only Food and Drug Administration (FDA)-approved drug in place for stroke patients, tissue plasminogen activator (tPA), has a rigid therapeutic window, closing at approximately 4.5 h after stroke onset. Due to this short time frame and other restrictions, such as any condition that increases a patient's risk for hemorrhaging, it has been predicted that <5% of ischemic stroke patients benefit from tPA. Given that rehabilitation therapy remains the only other option for stroke victims, there is a clear unmet clinical need for treatment available for the remaining 95%. While still considered an experimental treatment, the utilization of stem cell therapies for stroke holds consistent promise. Copious preclinical studies report the capacity for transplanted stem cells to rescue the brain parenchyma surrounding the stroke-induced infarct core. At present, the exact mechanisms in which stem cells contribute a robust therapeutic benefit remains unclear. Following stem cell administration, researchers have observed cell replacement, an increase in growth factors, and a reduction in inflammation. With a deeper understanding of the precise mechanism of stem cells, these therapies can be optimized in the clinic to afford the greatest therapeutic benefit. Recent studies have depicted a unique method of endogenous stem cell activation as a result of stem cell therapy. In both traumatic brain injury and stroke models, transplanted mesenchymal stromal cells (MSCs) facilitated a pathway between the neurogenic niches of the brain and the damaged area through extracellular matrix remodeling. The biobridge pioneered by the MSCs was utilized by the endogenous stem cells, and these cells were able to travel to the damaged areas distal to the neurogenic niches, a feat unachievable without prior remodeling. These studies broaden our understanding of stem cell interactions within the injured brain and help to guide both researchers and clinicians in developing an effective stem cell treatment for stroke. This paper is a review article. Referred literature in this paper has been listed in the references section. The datasets supporting the conclusions of this article are available online by searching various databases, including PubMed. Some original points in this article come from the laboratory practice in our research center and the authors’ experiences.
Collapse
Affiliation(s)
- Marci G Crowley
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | - Naoki Tajiri
- Department of Psychology, Graduate School of Psychology, Kibi International University, 8 Iga-machi, Takahashi-City, Okayama 716-8508, Japan
| |
Collapse
|
20
|
Jeon HJ, Park J, Shin JH, Chang MS. Insulin-like growth factor binding protein-6 released from human mesenchymal stem cells confers neuronal protection through IGF-1R-mediated signaling. Int J Mol Med 2017; 40:1860-1868. [PMID: 29039467 PMCID: PMC5716453 DOI: 10.3892/ijmm.2017.3173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/04/2017] [Indexed: 01/01/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hMSCs) are a desirable cell source for cell-based therapy to treat nervous system injuries due to their ability to differentiate into specific cell types. In addition to their multi-potency, hMSCs render the tissue microenvironment more favorable for tissue repair by secreting various growth factors. Our previous study demonstrated that hMSCs secrete several growth factors, including several insulin-like growth factor binding proteins (IGFBPs). Among these, IGFBP-6 binds with high affinity and inhibits insulin growth factor-2 (IGF-2) to inhibit the growth of IGF-2-dependent tumors. However, the function of IGFBP-6 in the nervous system remains to be fully elucidated. The present study investigated the protective effects of IGFBP-6 secreted by hMSCs on H2O2-injured primary cortical neuron cultures and lysolecithin-injured organotypic spinal cord slice cultures. Treatment of the H2O2-injured cortical neurons with conditioned media from hMSCs (hMSC-CM) increased the phosphorylation of Akt, reduced cell death and mitochondrial translocation of Bax, and regulated extracellular levels of IGF-1 and IGF-2. MTT assay, western blot analysis and ELISA were used to detect the cell viability and protein expression levels, respectively. An inhibitory antibody against IGFBP-6 eliminated this hMSC-CM-mediated neuroprotective effect in the injured cortical neuron cultures and spinal cord slice cultures. In addition, treatment with cyclolignan picropodophyllin, an inhibitor of IGF-1 receptor (IGF-1R), significantly inhibited neuronal protection by hMSC-CM. These findings demonstrated that hMSC-CM-mediated neuroprotection was attributed to IGF-1R-mediated signaling, potentiated via the inhibition of IGF-2 by IGFBP-6. The results of the present study provide insight into the mechanism by which hMSC administration may promote recovery from nerve injury.
Collapse
Affiliation(s)
- Hyo-Jin Jeon
- Laboratory of Stem Cell and Neurobiology, Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Jihye Park
- Laboratory of Stem Cell and Neurobiology, Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Joo-Hyun Shin
- Laboratory of Stem Cell and Neurobiology, Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Mi-Sook Chang
- Laboratory of Stem Cell and Neurobiology, Department of Oral Anatomy, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
21
|
Liang L, Li X, Li D, Jiang W, Wang H, Chen J, Sun Z, Zhang N, Zhu Y. The characteristics of stem cells in human degenerative intervertebral disc. Medicine (Baltimore) 2017; 96:e7178. [PMID: 28640098 PMCID: PMC5484206 DOI: 10.1097/md.0000000000007178] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 05/18/2017] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The aim of this study is to identify which possessed the best stem-cell-like characteristics in 3 kinds of cell in human degenerative intervertebral disc: NPSCs (nucleus pulposus-derived stem cells), AFSCs (annulus fibrosus-derived stem cells), or CESCs (cartilage endplate-derived stem cells). METHODS We separated the disc samples obtained from 15 surgically treated patients with disc degenerative diseases into nucleus pulposus, annulus fibrosus, and cartilage endplate. After cultivating, we used the cell counting kit-8 to analysis the cell activity of 3 kinds of disc tissue-derived stem cell separately; different stem cells were defined with multilineage (osteogenic, chondrogenic, and adipogenic) differentiation. We extracted the total RNA and detected the expression of different lineage differentiation-related genes with the real-time polymerase chain reaction (RT-PCR). RESULTS Cell morphology of NPSCs, AFSCs, and CESCs did not show significant difference. Cell proliferation capacity of NPSCs and AFSCs was stronger than that of CESCs. The differentiation outcomes showed that osteocyte-like cells were stained red by Alizarin red S, chondrocyte-like cells blue by toluidine blue, and adipocyte-like red by oil red O. The RT-PCR reflected that the expression of different lineage differentiation-related genes of AFSCs was stronger than NPSCs and CESCs. CONCLUSION In conclusion, we found that the cell morphology was not significantly different among NPSCs, AFSCs, and CESCs. Both differentiation and RT-PCR tests demonstrated that AFSCs had the best stem-cell-like characteristics in the human degenerative intervertebral disc.
Collapse
Affiliation(s)
- Lin Liang
- Department of Orthopaedics, Shangyu People's Hospital, Shaoxing
| | - Xuefeng Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou
| | - Dapeng Li
- Department of Orthopaedics, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Weimin Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou
| | - Heng Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou
| | - Jie Chen
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou
| | - Zhiyong Sun
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou
| | - Niannian Zhang
- Department of Orthopaedics, Shangyu People's Hospital, Shaoxing
| | - Yangyi Zhu
- Department of Orthopaedics, Shangyu People's Hospital, Shaoxing
| |
Collapse
|
22
|
Lee JY, Xu K, Nguyen H, Guedes VA, Borlongan CV, Acosta SA. Stem Cell-Induced Biobridges as Possible Tools to Aid Neuroreconstruction after CNS Injury. Front Cell Dev Biol 2017; 5:51. [PMID: 28540289 PMCID: PMC5424542 DOI: 10.3389/fcell.2017.00051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 04/21/2017] [Indexed: 12/12/2022] Open
Abstract
Notch-induced mesenchymal stromal cells (MSCs) mediate a distinct mechanism of repair after brain injury by forming a biobridge that facilitates biodistribution of host cells from a neurogenic niche to the area of injury. We have observed the biobridge in an area between the subventricular zone and the injured cortex using immunohistochemistry and laser capture. Cells in the biobridge express high levels of extracellular matrix metalloproteinases (MMPs), specifically MMP-9, which co-localized with a trail of MSCs graft. The transplanted stem cells then become almost undetectable, being replaced by newly recruited host cells. This stem cell-paved biobridge provides support for distal migration of host cells from the subventricular zone to the site of injury. Biobridge formation by transplanted stem cells seems to have a fundamental role in initiating endogenous repair processes. Two major stem cell-mediated repair mechanisms have been proposed thus far: direct cell replacement by transplanted grafts and bystander effects through the secretion of trophic factors including fibroblast growth factor 2 (FGF-2), epidermal growth factor (EGF), stem cell factor (SCF), erythropoietin, and brain-derived neurotrophic factor (BDNF) among others. This groundbreaking observation of biobridge formation by transplanted stem cells represents a novel mechanism for stem cell mediated brain repair. Future studies on graft-host interaction will likely establish biobridge formation as a fundamental mechanism underlying therapeutic effects of stem cells and contribute to the scientific pursuit of developing safe and efficient therapies not only for traumatic brain injury but also for other neurological disorders. The aim of this review is to hypothetically extend concepts related to the formation of biobridges in other central nervous system disorders.
Collapse
Affiliation(s)
- Jea Y Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Kaya Xu
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Hung Nguyen
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Vivian A Guedes
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida College of MedicineTampa, FL, USA
| |
Collapse
|
23
|
Beltz BS, Brenneis G, Benton JL. Adult Neurogenesis: Lessons from Crayfish and the Elephant in the Room. BRAIN, BEHAVIOR AND EVOLUTION 2016; 87:146-155. [DOI: 10.1159/000447084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The 1st-generation neural precursors in the crustacean brain are functionally analogous to neural stem cells in mammals. Their slow cycling, migration of their progeny, and differentiation of their descendants into neurons over several weeks are features of the neural precursor lineage in crayfish that also characterize adult neurogenesis in mammals. However, the 1st-generation precursors in crayfish do not self-renew, contrasting with conventional wisdom that proposes the long-term self-renewal of adult neural stem cells. Nevertheless, the crayfish neurogenic niche, which contains a total of 200-300 cells, is never exhausted and neurons continue to be produced in the brain throughout the animal's life. The pool of neural precursors in the niche therefore cannot be a closed system, and must be replenished from an extrinsic source. Our in vitro and in vivo data show that cells originating in the innate immune system (but not other cell types) are attracted to and incorporated into the neurogenic niche, and that they express a niche-specific marker, glutamine synthetase. Further, labeled hemocytes that undergo adoptive transfer to recipient crayfish generate cells in neuronal clusters in the olfactory pathway of the adult brain. These hemocyte descendants express appropriate neurotransmitters and project to target areas typical of neurons in these regions. These studies indicate that under natural conditions, the immune system provides neural precursors supporting adult neurogenesis in the crayfish brain, challenging the canonical view that ectodermal tissues generating the embryonic nervous system are the sole source of neurons in the adult brain. However, these are not the first studies that directly implicate the immune system as a source of neural precursor cells. Several types of data in mammals, including adoptive transfers of bone marrow or stem cells as well as the presence of fetal microchimerism, suggest that there must be a population of cells that are able to access the brain and generate new neurons in these species.
Collapse
|
24
|
Zhou L, Chen X, Liu T, Gong Y, Chen S, Pan G, Cui W, Luo ZP, Pei M, Yang H, He F. Melatonin reverses H2 O2 -induced premature senescence in mesenchymal stem cells via the SIRT1-dependent pathway. J Pineal Res 2015; 59:190-205. [PMID: 25975679 PMCID: PMC4523475 DOI: 10.1111/jpi.12250] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/08/2015] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) represent an attractive source for stem cell-based regenerative therapy, but they are vulnerable to oxidative stress-induced premature senescence in pathological conditions. We previously reported antioxidant and antiarthritic effects of melatonin on MSCs against proinflammatory cytokines. In this study, we hypothesized that melatonin could protect MSCs from premature senescence induced by hydrogen peroxide (H2 O2 ) via the silent information regulator type 1 (SIRT1)-dependent pathway. In response to H2 O2 at a sublethal concentration of 200 μm, human bone marrow-derived MSCs (BM-MSCs) underwent growth arrest and cellular senescence. Treatment with melatonin before H2 O2 exposure cannot significantly prevent premature senescence; however, treatment with melatonin subsequent to H2 O2 exposure successfully reversed the senescent phenotypes of BM-MSCs in a dose-dependent manner. This result was made evident by improved cell proliferation, decreased senescence-associated β-galactosidase activity, and the improved entry of proliferating cells into the S phase. In addition, treatment with 100 μm melatonin restored the osteogenic differentiation potential of BM-MSCs that was inhibited by H2 O2 -induced premature senescence. We also found that melatonin attenuated the H2 O2 -stimulated phosphorylation of p38 mitogen-activated protein kinase, decreased expression of the senescence-associated protein p16(INK) (4α) , and increased SIRT1. Further molecular experiments revealed that luzindole, a nonselective antagonist of melatonin receptors, blocked melatonin-mediated antisenescence effects. Inhibition of SIRT1 by sirtinol counteracted the protective effects of melatonin, suggesting that melatonin reversed the senescence in cells through the SIRT1-dependent pathway. Together, these findings lay new ground for understanding oxidative stress-induced premature senescence and open perspectives for therapeutic applications of melatonin in stem cell-based regenerative medicine.
Collapse
Affiliation(s)
- Long Zhou
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xi Chen
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
- School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, China
| | - Tao Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yihong Gong
- School of Engineering, Sun Yat-sen University, Guangzhou, China
| | - Sijin Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoqing Pan
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenguo Cui
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zong-Ping Luo
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, USA
| | - Huilin Yang
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Sala E, Genua M, Petti L, Anselmo A, Arena V, Cibella J, Zanotti L, D'Alessio S, Scaldaferri F, Luca G, Arato I, Calafiore R, Sgambato A, Rutella S, Locati M, Danese S, Vetrano S. Mesenchymal Stem Cells Reduce Colitis in Mice via Release of TSG6, Independently of Their Localization to the Intestine. Gastroenterology 2015; 149:163-176.e20. [PMID: 25790743 DOI: 10.1053/j.gastro.2015.03.013] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 02/19/2015] [Accepted: 03/06/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Mesenchymal stem cells (MSCs) are pluripotent cells that can promote expansion of immune regulatory cells and might be developed for the treatment of immune disorders, including inflammatory bowel diseases. MSCs were reported to reduce colitis in mice; we investigated whether MSC localization to the intestine and production of paracrine factors, including tumor necrosis factor-induced protein 6 (TSG6), were required for these effects. METHODS MSCs were isolated from bone marrow (BM-MSCs) of 4- to 6-week-old C57BL/6, C57BL/6-green fluorescent protein, or Balb/c Tsg6-/- male mice. Colitis was induced by ad libitum administration of dextran sulfate sodium for 10 days; after 5 days the mice were given intraperitoneal injections of BM-MSCs or saline (controls). Blood samples and intestinal tissues were collected 24, 48, 96, and 120 hours later; histologic and flow cytometry analyses were performed. RESULTS Injection of BM-MSCs reduced colitis in mice, increasing body weight and reducing markers of intestinal inflammation, compared with control mice. However, fewer than 1% of MSCs reached the inflamed colon. Most of the BM-MSCs formed aggregates in the peritoneal cavity. The aggregates contained macrophages and B and T cells, and produced immune-regulatory molecules including FOXP3, interleukin (IL)10, transforming growth factor-β, arginase type II, chemokine (C-C motif) ligand 22 (CCL22), heme oxygenase-1, and TSG6. Serum from mice given BM-MSCs, compared with mice given saline, had increased levels of TSG6. Injection of TSG6 reduced the severity of colitis in mice, along with the numbers of CD45+ cells, neutrophils and metalloproteinase activity in the mucosa, while increasing the percentage of Foxp3CD45+ cells. TSG6 injection also promoted the expansion of regulatory macrophages that expressed IL10 and inducible nitric oxide synthase, and reduced serum levels of interferon-γ, IL6, and tumor necrosis factor. Tsg6-/- MSCs did not suppress the mucosal inflammatory response in mice with colitis. CONCLUSIONS BM-MSCs injected into mice with colitis do not localize to the intestine but instead form aggregates in the peritoneum where they produce immunoregulatory molecules, including TSG6, that reduce intestinal inflammation. TSG6 is sufficient to reduce intestinal inflammation in mice with colitis.
Collapse
Affiliation(s)
- Emanuela Sala
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Marco Genua
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Luciana Petti
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Achille Anselmo
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Vincenzo Arena
- Department of Pathology, Catholic University of Rome, Rome, Italy
| | - Javier Cibella
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Lucia Zanotti
- Department of Immunology, Humanitas Clinical and Research Center, Milan, Italy
| | - Silvia D'Alessio
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Franco Scaldaferri
- Internal Medicine, Gastroenterology Division, Catholic University of the Sacred Heart, Rome, Italy
| | - Giovanni Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Alessandro Sgambato
- Institute of General Pathology, Catholic University of the Sacred Heart, Rome, Italy
| | - Sergio Rutella
- Clinical Research Center, Division of Translational Medicine, Sidra Medical & Research Center, Doha, Qatar
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Silvio Danese
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy
| | - Stefania Vetrano
- Inflammatory Bowel Disease Center, Humanitas Clinical and Research Center, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Milan, Italy.
| |
Collapse
|
26
|
Therapeutic effectiveness of bone marrow-derived mesenchymal stem cell administration against acute pulmonary thromboembolism in a mouse model. Thromb Res 2015; 135:990-9. [DOI: 10.1016/j.thromres.2015.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 01/31/2015] [Accepted: 02/08/2015] [Indexed: 02/04/2023]
|
27
|
Zou Z, Cai Y, Chen Y, Chen S, Liu L, Shen Z, Zhang S, Xu L, Chen Y. Bone marrow-derived mesenchymal stem cells attenuate acute liver injury and regulate the expression of fibrinogen-like-protein 1 and signal transducer and activator of transcription 3. Mol Med Rep 2015; 12:2089-97. [PMID: 25901902 DOI: 10.3892/mmr.2015.3660] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/27/2015] [Indexed: 01/16/2023] Open
Abstract
In recent years, bone marrow-derived mesenchymal stem cells (BMSCs) have been demonstrated to exert extensive therapeutic effects on acute liver injury; however, the underlying mechanisms of these effects have remained to be elucidated. The present study focused on the potential anti-apoptotic and pro-regenerative effects of BMSCs in D-galactosamine (D-Gal) and lipopolysaccharide (LPS)-induced acute liver injury in rats. An experimental rat acute liver injury model was established by intraperitoneal injection of D-Gal (400 mg/kg) and LPS (80 μg/kg). BMSCs and an identical volume of saline were administered via the caudal vein 2 h after the D-Gal and LPS challenge. Subsequently, the serum samples were collected to detect the levels of alanine aminotransferase and aspartate aminotransferase. Hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated nick-end labeling assay and immunohistochemical staining were performed to determine apoptosis, regeneration and histological changes of liver sections. Western blotting and reverse transcription-quantitative polymerase chain reaction were performed to detect the protein and mRNA expression levels of fibrinogen-like-protein 1 (FGL1), phosphorylated signal transducer and activator of transcription 3 (p-STAT3), STAT3 and B-cell lymphoma 2 (Bcl-2) and Bcl-2 associated X protein (Bax) in liver tissue samples. The results indicated that intravenous transplantation of BMSCs significantly decreased the levels of alanine aminotransferase and aspartate aminotransferase, and reduced hepatocellular necrosis and inflammatory cell infiltration. Additionally, a terminal deoxynucleotidyl transferase-mediated nick-end labeling assay and immunohistochemical staining revealed that BMSC treatment reduced hepatocyte apoptosis and enhanced liver regeneration. Furthermore, Bcl-2 expression was increased, whilst the protein expression of Bax was reduced. The expression of FGL1 and p-STAT3 were elevated concurrently with the improvement of liver function. These results demonstrated that BMSCs may provide a promising potential agent for the prevention of acute liver injury via inhibition of hepatocyte apoptosis and acceleration of liver regeneration. The mechanism may be, a least in part, a consequence of the upregulation of FGL1 expression and the induction of STAT3 phosphorylation.
Collapse
Affiliation(s)
- Zhuolin Zou
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yijing Cai
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yi Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Si Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Liyuan Liu
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhonghai Shen
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, P.R. China
| | - Sainan Zhang
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lanman Xu
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yongping Chen
- Department of Infectious Disease, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
28
|
Duncan K, Gonzales-Portillo GS, Acosta SA, Kaneko Y, Borlongan CV, Tajiri N. Stem cell-paved biobridges facilitate stem transplant and host brain cell interactions for stroke therapy. Brain Res 2015; 1623:160-5. [PMID: 25770817 DOI: 10.1016/j.brainres.2015.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/23/2015] [Accepted: 03/03/2015] [Indexed: 01/01/2023]
Abstract
Distinguished by an infarct core encased within a penumbra, stroke remains a primary source of mortality within the United States. While our scientific knowledge regarding the pathology of stroke continues to improve, clinical treatment options for patients suffering from stroke are extremely limited. Tissue plasminogen activator (tPA) remains the sole FDA-approved drug proven to be helpful following stroke. However, due to the need to administer the drug within 4.5h of stroke onset its usefulness is constrained to less than 5% of all patients suffering from ischemic stroke. One experimental therapy for the treatment of stroke involves the utilization of stem cells. Stem cell transplantation has been linked to therapeutic benefit by means of cell replacement and release of growth factors; however the precise means by which this is accomplished has not yet been clearly delineated. Using a traumatic brain injury model, we recently demonstrated the ability of transplanted mesenchymal stromal cells (MSCs) to form a biobridge connecting the area of injury to the neurogenic niche within the brain. We hypothesize that MSCs may also have the capacity to create a similar biobridge following stroke; thereby forming a conduit between the neurogenic niche and the stroke core and peri-infarct area. We propose that this biobridge could assist and promote interaction of host brain cells with transplanted stem cells and offer more opportunities to enhance the effectiveness of stem cell therapy in stroke. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Kelsey Duncan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States
| | - Gabriel S Gonzales-Portillo
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States
| | - Sandra A Acosta
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States
| | - Yuji Kaneko
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States.
| | - Naoki Tajiri
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd, Tampa, FL 33612, United States.
| |
Collapse
|
29
|
Bone Marrow-Derived Mesenchymal Stem Cells Repair Necrotic Pancreatic Tissue and Promote Angiogenesis by Secreting Cellular Growth Factors Involved in the SDF-1 α /CXCR4 Axis in Rats. Stem Cells Int 2015; 2015:306836. [PMID: 25810724 PMCID: PMC4355908 DOI: 10.1155/2015/306836] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 12/29/2014] [Accepted: 01/08/2015] [Indexed: 01/08/2023] Open
Abstract
Acute pancreatitis (AP), a common acute abdominal disease, 10%-20% of which can evolve into severe acute pancreatitis (SAP), is of significant morbidity and mortality. Bone marrow-derived mesenchymal stem cells (BMSCs) have been reported to have a potential therapeutic role on SAP, but the specific mechanism is unclear. Therefore, we conducted this experiment to shed light on the probable mechanism. We validated that SDF-1α significantly stimulated the expressions of VEGF, ANG-1, HGF, TGF-β, and CXCR4 in BMSCs, which were inhibited by its receptor agonist, AMD3100. The capacities of proliferation, migration, and repair of human umbilical vein endothelial cells were enhanced by BMSCs supernatant. Meanwhile, BMSCs supernatant could also promote angiogenesis, especially after the stimulation with SDF-1α. In vivo, the migration of BMSCs was regulated by SDF-1α/CXCR4 axis. Moreover, transplanted BMSCs could significantly alleviate SAP, reduce the systematic inflammation (TNF-α↓, IL-1β↓, IL-6↓, IL-4↑, IL-10↑, and TGF-β↑), and promote tissue repair and angiogenesis (VEGF↑, ANG-1↑, HGF↑, TGF-β↑, and CD31↑), compared with the SAP and anti-CXCR4 groups. Taken together, the results showed that BMSCs ameliorated SAP and the SDF-1α/CXCR4 axis was involved in the repair and regeneration process.
Collapse
|
30
|
Chang KA, Lee JH, Suh YH. Therapeutic potential of human adipose-derived stem cells in neurological disorders. J Pharmacol Sci 2014; 126:293-301. [PMID: 25409785 DOI: 10.1254/jphs.14r10cp] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Stem cell therapy has been noted as a novel strategy to various diseases including neurological disorders such as Alzheimer's disease, Parkinson's disease, stroke, amyotrophic lateral sclerosis, and Huntington's disease that have no effective treatment available to date. The adipose-derived stem cells (ASCs), mesenchymal stem cells (MSCs) isolated from adipose tissue, are well known for their pluripotency with the ability to differentiate into various types of cells and immuno-modulatory property. These biological features make ASCs a promising source for regenerative cell therapy in neurological disorders. Here we discuss the recent progress of regenerative therapies in various neurological disorders utilizing ASCs.
Collapse
Affiliation(s)
- Keun-A Chang
- Department of Pharmacology, College of Medicine, Neuroscience Research Institute, Gachon University, Korea
| | | | | |
Collapse
|
31
|
Benton JL, Kery R, Li J, Noonin C, Söderhäll I, Beltz BS. Cells from the immune system generate adult-born neurons in crayfish. Dev Cell 2014; 30:322-33. [PMID: 25117683 DOI: 10.1016/j.devcel.2014.06.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Revised: 04/08/2014] [Accepted: 06/19/2014] [Indexed: 10/24/2022]
Abstract
Neurogenesis is an ongoing process in the brains of adult decapod crustaceans. However, the first-generation precursors that produce adult-born neurons, which reside in a neurogenic niche, are not self-renewing in crayfish and must be replenished. The source of these neuronal precursors is unknown. Here, we report that adult-born neurons in crayfish can be derived from hemocytes. Following adoptive transfer of 5-ethynyl-2'-deoxyuridine (EdU)-labeled hemocytes, labeled cells populate the neurogenic niche containing the first-generation neuronal precursors. Seven weeks after adoptive transfer, EdU-labeled cells are located in brain clusters 9 and 10 (where adult-born neurons differentiate) and express appropriate neurotransmitters. Moreover, the number of cells composing the neurogenic niche in crayfish is tightly correlated with total hemocyte counts (THCs) and can be manipulated by raising or lowering THC. These studies identify hemocytes as a source of adult-born neurons in crayfish and demonstrate that the immune system is a key contributor to adult neurogenesis.
Collapse
Affiliation(s)
- Jeanne L Benton
- Neuroscience Program, Wellesley College, Wellesley, MA 02481, USA
| | - Rachel Kery
- Neuroscience Program, Wellesley College, Wellesley, MA 02481, USA
| | - Jingjing Li
- Neuroscience Program, Wellesley College, Wellesley, MA 02481, USA
| | - Chadanat Noonin
- Department of Comparative Physiology, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Irene Söderhäll
- Department of Comparative Physiology, Uppsala University, SE-752 36 Uppsala, Sweden.
| | - Barbara S Beltz
- Neuroscience Program, Wellesley College, Wellesley, MA 02481, USA.
| |
Collapse
|
32
|
Abstract
Adult neurogenic niches are present in both vertebrates and invertebrates. Where do stem cells populating these niches originate, and what are the mechanisms maintaining their self-renewal? In this issue of Developmental Cell, Benton et al. (2014) show that in crayfish, hemolymph-derived cells enter a neurogenic niche to replenish neural progenitors.
Collapse
|
33
|
Paradisi M, Alviano F, Pirondi S, Lanzoni G, Fernandez M, Lizzo G, Giardino L, Giuliani A, Costa R, Marchionni C, Bonsi L, Calzà L. Human Mesenchymal Stem Cells Produce Bioactive Neurotrophic Factors: Source, Individual Variability and Differentiation Issues. Int J Immunopathol Pharmacol 2014; 27:391-402. [DOI: 10.1177/039463201402700309] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The possible use of cell therapies for neurological lesions and disorders is regarded as a very promising strategy. However, many issues related to cell type, tissue donor, expected biological action etc., are still open. In this study human mesenchymal stem cells derived from different fetal and adult tissues were examined in order to explore growth and neurotrophic factor synthesis and biological action, also considering the individual variability of the donors. Cells were derived from different human tissues and characterized according to the guidelines of the International Society for Cellular Therapy. Growth and neurotrophic factor synthesis was evaluated by real time PCR, biological assays and ELISA. It was found that human mesenchymal stem cells produce vascular endothelial-, nerve-growth factor (VEGF, NGF), brain-derived-, ciliary- and glial-derived neurotrophic factors (BDNF, CDGF, GDNF), which are neuroprotective molecules, but the source and the donor influence the synthesis rate. Accordingly, it is suggested that the source and the individual variability are key issues to be considered in the perspective of the clinical use of mesenchymal stem cells in neurological disorders.
Collapse
Affiliation(s)
- M. Paradisi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - F. Alviano
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - S. Pirondi
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - G. Lanzoni
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - M. Fernandez
- Health Science and Technology Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| | - G. Lizzo
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - L. Giardino
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
- Health Science and Technology Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| | - A. Giuliani
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - R. Costa
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - C. Marchionni
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - L. Bonsi
- Department of Experimental, Diagnostic and Specialty Medicine, Unit of Histology, Embryology and Applied Biology, University of Bologna, Bologna, Italy
| | - L. Calzà
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
- Health Science and Technology Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| |
Collapse
|
34
|
Tajiri N, Duncan K, Antoine A, Pabon M, Acosta SA, de la Pena I, Hernadez-Ontiveros DG, Shinozuka K, Ishikawa H, Kaneko Y, Yankee E, McGrogan M, Case C, Borlongan CV. Stem cell-paved biobridge facilitates neural repair in traumatic brain injury. Front Syst Neurosci 2014; 8:116. [PMID: 25009475 PMCID: PMC4068001 DOI: 10.3389/fnsys.2014.00116] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 05/28/2014] [Indexed: 12/18/2022] Open
Abstract
Modified mesenchymal stromal cells (MSCs) display a unique mechanism of action during the repair phase of traumatic brain injury by exhibiting the ability to build a biobridge between the neurogenic niche and the site of injury. Immunohistochemistry and laser capture assay have visualized this biobridge in the area between the neurogenic subventricular zone and the injured cortex. This biobridge expresses high levels of extracellular matrix metalloproteinases (MMPs), which are initially co-localized with a stream of transplanted MSCs, but later this region contains only few to non-detectable grafts and becomes overgrown by newly recruited host cells. We have reported that long-distance migration of host cells from the neurogenic niche to the injured brain site can be attained via these transplanted stem cell-paved biobridges, which serve as a key regenerative process for the initiation of endogenous repair mechanisms. Thus, far the two major schools of discipline in stem cell repair mechanisms support the idea of "cell replacement" and the bystander effects of "trophic factor secretion." Our novel observation of stem cell-paved biobridges as pathways for directed migration of host cells from neurogenic niche toward the injured brain site adds another mode of action underlying stem cell therapy. More in-depth investigations on graft-host interaction will likely aid translational research focused on advancing this stem cell-paved biobridge from its current place, as an equally potent repair mechanism as cell replacement and trophic factor secretion, into a new treatment strategy for traumatic brain injury and other neurological disorders.
Collapse
Affiliation(s)
- Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Kelsey Duncan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Alesia Antoine
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Mibel Pabon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Sandra A Acosta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Ike de la Pena
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Diana G Hernadez-Ontiveros
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Kazutaka Shinozuka
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Hiroto Ishikawa
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | | | | | | | - Cesar V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| |
Collapse
|
35
|
Tajiri N, Kaneko Y, Shinozuka K, Ishikawa H, Yankee E, McGrogan M, Case C, Borlongan CV. Stem cell recruitment of newly formed host cells via a successful seduction? Filling the gap between neurogenic niche and injured brain site. PLoS One 2013; 8:e74857. [PMID: 24023965 PMCID: PMC3762783 DOI: 10.1371/journal.pone.0074857] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/06/2013] [Indexed: 01/24/2023] Open
Abstract
Here, we report that a unique mechanism of action exerted by stem cells in the repair of the traumatically injured brain involves their ability to harness a biobridge between neurogenic niche and injured brain site. This biobridge, visualized immunohistochemically and laser captured, corresponded to an area between the neurogenic subventricular zone and the injured cortex. That the biobridge expressed high levels of extracellular matrix metalloproteinases characterized initially by a stream of transplanted stem cells, but subsequently contained only few to non-detectable grafts and overgrown by newly formed host cells, implicates a novel property of stem cells. The transplanted stem cells manifest themselves as pathways for trafficking the migration of host neurogenic cells, but once this biobridge is formed between the neurogenic site and the injured brain site, the grafted cells disappear and relinquish their task to the host neurogenic cells. Our findings reveal that long-distance migration of host cells from the neurogenic niche to the injured brain site can be achieved through transplanted stem cells serving as biobridges for initiation of endogenous repair mechanisms. This is the first report of a stem cell-paved “biobridge”. Indeed, to date the two major schools of discipline in stem cell repair mechanism primarily support the concept of “cell replacement” and bystander effects of “trophic factor secretion”. The present novel observations of a stem cell seducing a host cell to engage in brain repair advances basic science concepts on stem cell biology and extracellular matrix, as well as provokes translational research on propagating this stem cell-paved biobridge beyond cell replacement and trophic factor secretion for the treatment of traumatic brain injury and other neurological disorders.
Collapse
Affiliation(s)
- Naoki Tajiri
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Kazutaka Shinozuka
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Hiroto Ishikawa
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
| | - Ernest Yankee
- Sanbio Inc, Mountain View, California, United States of America
| | | | - Casey Case
- Sanbio Inc, Mountain View, California, United States of America
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, Florida, United States of America
- * E-mail:
| |
Collapse
|
36
|
ZHAO MANMAN, CUI JIANZHONG, CUI YING, LI RAN, TIAN YANXIA, SONG SIXIN, ZHANG JUAN, GAO JUNLING. Therapeutic effect of exogenous bone marrow-derived mesenchymal stem cell transplantation on silicosis via paracrine mechanisms in rats. Mol Med Rep 2013; 8:741-6. [DOI: 10.3892/mmr.2013.1580] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 04/09/2013] [Indexed: 01/08/2023] Open
|
37
|
Noda Y, Nishizaki K, Yoshinobu J, Orita Y, Tsujigiwa H, Yamada M. The engraftment and differentiation of transplanted bone marrow-derived cells in the olfactory bulb after methimazole administration. Acta Otolaryngol 2013; 133:951-6. [PMID: 23822107 DOI: 10.3109/00016489.2013.803153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONCLUSION Bone marrow-derived cells can be engrafted in the olfactory bulb and a few cells can differentiate into mitral/tufted cells in the olfactory bulb. OBJECTIVES To investigate whether bone marrow-derived cells can be engrafted into the olfactory bulb and differentiate into neurons and glial cells after methimazole administration. METHODS Bone marrow of GFP (green fluorescence protein) mice was transplanted into lethally irradiated recipient mice. Immunostaining was performed to confirm the cell types of bone marrow-derived cells expressing GFP. RESULTS GFP-positive cells were observed in the olfactory bulb at 2 days after methimazole administration. The number of dendritic GFP-positive cells increased up to 30 days after methimazole administration and then decreased. Double immunostaining for GFP and Iba1 or TBX21 showed that a large population of the GFP-positive cells had characteristics of microglia/macrophages and a few cells had characteristics of mitral/tufted cells.
Collapse
Affiliation(s)
- Yohei Noda
- Department of Otolaryngology-Head & Neck Surgery, Okayama, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Grafi G. Stress cycles in stem cells/iPSCs development: implications for tissue repair. Biogerontology 2013; 14:603-8. [PMID: 23852045 DOI: 10.1007/s10522-013-9445-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 07/07/2013] [Indexed: 12/12/2022]
Abstract
Stem cells have become a major topic, both publicly and scientifically, owing to their potential to cure diseases and repair damaged tissues. Particular attention has been given to the so-called "induced pluripotent stem cells" (iPSCs) in which somatic cells are induced by the expression of transcription factor encoding transgenes-a methodology first established by Takahashi and Yamanaka (Cell 126:663-676, 2006)-to acquire pluripotent state. This methodology has captured researchers' imagination as a potential procedure to obtain patient-specific therapies while also solving both the problem of transplant rejection and the ethical concerns often raised regarding the use of embryonic stem cells in regenerative medicine. The study of the biology of stem cells/iPSCs, in recent years, has uncovered some fundamental weaknesses that undermine their potential use in transplantation therapies.
Collapse
Affiliation(s)
- Gideon Grafi
- French Associates Institute for Agriculture and Biotechnology of Drylands, Jacob Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, 84990, Midreshet Ben-Gurion, Israel,
| |
Collapse
|
39
|
Zanotti L, Sarukhan A, Dander E, Castor M, Cibella J, Soldani C, Trovato AE, Ploia C, Luca G, Calvitti M, Mancuso F, Arato I, Golemac M, Jonjic N, Biondi A, Calafiore R, Locati M, D'Amico G, Viola A. Encapsulated mesenchymal stem cells for in vivo immunomodulation. Leukemia 2012; 27:500-3. [PMID: 22878604 DOI: 10.1038/leu.2012.202] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
40
|
Uccelli A, Milanese M, Principato MC, Morando S, Bonifacino T, Vergani L, Giunti D, Voci A, Carminati E, Giribaldi F, Caponnetto C, Bonanno G. Intravenous mesenchymal stem cells improve survival and motor function in experimental amyotrophic lateral sclerosis. Mol Med 2012; 18:794-804. [PMID: 22481270 DOI: 10.2119/molmed.2011.00498] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2011] [Accepted: 03/30/2012] [Indexed: 12/13/2022] Open
Abstract
Despite some advances in the understanding of amyotrophic lateral sclerosis (ALS) pathogenesis, significant achievements in treating this disease are still lacking. Mesenchymal stromal (stem) cells (MSCs) have been shown to be effective in several models of neurological disease. To determine the effects of the intravenous injection of MSCs in an ALS mouse model during the symptomatic stage of disease, MSCs (1 × 10⁶) were intravenously injected in mice expressing human superoxide dismutase 1 (SOD1) carrying the G93A mutation (SOD1/G93A) presenting with experimental ALS. Survival, motor abilities, histology, oxidative stress markers and [³H]D-aspartate release in the spinal cord were investigated. MSC injection in SOD1/G93A mice improved survival and motor functions compared with saline-injected controls. Injected MSCs scantly home to the central nervous system and poorly engraft. We observed a reduced accumulation of ubiquitin agglomerates and of activated astrocytes and microglia in the spinal cord of MSC-treated SOD1/G93A mice, with no changes in the number of choline acetyltransferase- and glutamate transporter type 1-positive cells. MSC administration turned around the upregulation of metallothionein mRNA expression and of the activity of the antioxidant enzyme glutathione S-transferase, both associated with disease progression. Last, we observed that MSCs reverted both spontaneous and stimulus-evoked neuronal release of [³H]D-aspartate, a marker of endogenous glutamate, which is upregulated in SOD1/G93A mice. These findings suggest that intravenous administration of MSCs significantly improves the clinical outcome and pathological scores of mutant SOD1/G93A mice, thus providing the rationale for their exploitation for the treatment of ALS.
Collapse
Affiliation(s)
- Antonio Uccelli
- Department of Neurosciences Ophthalmology and Genetics, University of Genoa, Genoa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sassoli C, Pini A, Chellini F, Mazzanti B, Nistri S, Nosi D, Saccardi R, Quercioli F, Zecchi-Orlandini S, Formigli L. Bone marrow mesenchymal stromal cells stimulate skeletal myoblast proliferation through the paracrine release of VEGF. PLoS One 2012; 7:e37512. [PMID: 22815682 PMCID: PMC3398011 DOI: 10.1371/journal.pone.0037512] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 04/24/2012] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are the leading cell candidates in the field of regenerative medicine. These cells have also been successfully used to improve skeletal muscle repair/regeneration; however, the mechanisms responsible for their beneficial effects remain to be clarified. On this basis, in the present study, we evaluated in a co-culture system, the ability of bone-marrow MSCs to influence C2C12 myoblast behavior and analyzed the cross-talk between the two cell types at the cellular and molecular level. We found that myoblast proliferation was greatly enhanced in the co-culture as judged by time lapse videomicroscopy, cyclin A expression and EdU incorporation. Moreover, myoblasts immunomagnetically separated from MSCs after co-culture expressed higher mRNA and protein levels of Notch-1, a key determinant of myoblast activation and proliferation, as compared with the single culture. Notch-1 intracellular domain and nuclear localization of Hes-1, a Notch-1 target gene, were also increased in the co-culture. Interestingly, the myoblastic response was mainly dependent on the paracrine release of vascular endothelial growth factor (VEGF) by MSCs. Indeed, the addition of MSC-derived conditioned medium (CM) to C2C12 cells yielded similar results as those observed in the co-culture and increased the phosphorylation and expression levels of VEGFR. The treatment with the selective pharmacological VEGFR inhibitor, KRN633, resulted in a marked attenuation of the receptor activation and concomitantly inhibited the effects of MSC-CM on C2C12 cell growth and Notch-1 signaling. In conclusion, this study provides novel evidence for a role of MSCs in stimulating myoblast cell proliferation and suggests that the functional interaction between the two cell types may be exploited for the development of new and more efficient cell-based skeletal muscle repair strategies.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Alessandro Pini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Flaminia Chellini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Benedetta Mazzanti
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Riccardo Saccardi
- Department of Hematology, Cord Blood Bank, Careggi Hospital, University of Florence, Florence, Italy
| | - Franco Quercioli
- National Institute of Optics (INO), National Research Council (CNR), Sesto Fiorentino, Florence, Italy
| | - Sandra Zecchi-Orlandini
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
| | - Lucia Formigli
- Department of Human Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy
- * E-mail:
| |
Collapse
|
42
|
Emmert MY, Emmert LS, Martens A, Ismail I, Schmidt-Richter I, Gawol A, Seifert B, Haverich A, Martin U, Gruh I. Higher frequencies of BCRP+ cardiac resident cells in ischaemic human myocardium. Eur Heart J 2012; 34:2830-8. [PMID: 22736676 DOI: 10.1093/eurheartj/ehs156] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AIMS Several cardiac resident progenitor cell types have been reported for the adult mammalian heart. Here we characterize their frequencies and distribution pattern in non-ischaemic human myocardial tissue and after ischaemic events. METHODS AND RESULTS We obtained 55 biopsy samples from human atria and ventricles and used immunohistological analysis to investigate two cardiac cell types, characterized by the expression of breast cancer resistance protein (BCRP)/ABCG2 [for side population (SP) cells] or c-kit. Highest frequencies of BCRP+ cells were detected in the ischaemic right atria with a median of 5.40% (range: 2.48-11.1%) vs. 4.40% (1.79-7.75%) in the non-ischaemic right atria (P = 0.47). Significantly higher amounts were identified in ischaemic compared with non-ischaemic ventricles, viz. 5.44% (3.24-9.30%) vs. 0.74% (0-5.23%) (P = 0.016). Few numbers of BCRP+ cells co-expressed the cardiac markers titin, sarcomeric α-actinin, or Nkx2.5; no co-expression of BCRP and progenitor cell marker Sca-1 or pluripotency markers Oct-3/4, SSEA-3, and SSEA-4 was detected. C-kit+ cells displayed higher frequencies in ischaemic (ratio: 1:25 000 ± 2500 of cell counts) vs. non-ischaemic myocardium (1:105 000 ± 43 000). Breast cancer resistance protein+/c-kit+ cells were not identified. Following in vitro differentiation, BCRP+ cells isolated from human heart biopsy samples (n = 6) showed expression of cardiac troponin T and α-myosin heavy-chain, but no full differentiation into functional beating cardiomyocytes was observed. CONCLUSION We were able to demonstrate that BCRP+/CD31- cells are more abundant in the heart than their c-kit+ counterparts. In the non-ischaemic hearts, they are preferentially located in the atria. Following ischaemia, their numbers are elevated significantly. Our data might provide a valuable snapshot at potential progenitor cells after acute ischaemia in vivo, and mapping of these easily accessible cells may influence future cell therapeutic strategies.
Collapse
Affiliation(s)
- Maximilian Y Emmert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, LEBAO and Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Cluster of Excellence REBIRTH, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
LI LI, WANG JING, TANG LING, YU XIN, SUI YI, ZHANG CHAODONG. Co-culture with bone marrow stromal cells protects PC12 neuronal cells from tumor necrosis factor-α-induced apoptosis by inhibiting the tumor necrosis factor receptor/caspase signaling pathway. Mol Med Rep 2012; 12:261-6. [DOI: 10.3892/mmr.2015.3421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 01/02/2015] [Indexed: 11/05/2022] Open
|
44
|
Cervelló I, Gil-Sanchis C, Mas A, Faus A, Sanz J, Moscardó F, Higueras G, Sanz MA, Pellicer A, Simón C. Bone marrow-derived cells from male donors do not contribute to the endometrial side population of the recipient. PLoS One 2012; 7:e30260. [PMID: 22276168 PMCID: PMC3261882 DOI: 10.1371/journal.pone.0030260] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/12/2011] [Indexed: 01/28/2023] Open
Abstract
Accumulated evidence demonstrates the existence of bone marrow-derived cells origin in the endometria of women undergoing bone marrow transplantation (BMT). In these reports, cells of a bone marrow (BM) origin are able to differentiate into endometrial cells, although their contribution to endometrial regeneration is not yet clear. We have previously demonstrated the functional relevance of side population (SP) cells as the endogenous source of somatic stem cells (SSC) in the human endometrium. The present work aims to understand the presence and contribution of bone marrow-derived cells to the endometrium and the endometrial SP population of women who received BMT from male donors. Five female recipients with spontaneous or induced menstruations were selected and their endometrium was examined for the contribution of XY donor-derived cells using fluorescent in situ hybridization (FISH), telomapping and SP method investigation. We confirm the presence of XY donor-derived cells in the recipient endometrium ranging from 1.7% to 2.62%. We also identify 0.45-0.85% of the donor-derived cells in the epithelial compartment displaying CD9 marker, and 1.0-1.83% of the Vimentin-positive XY donor-derived cells in the stromal compartment. Although the percentage of endometrial SP cells decreased, possibly being due to chemotherapy applied to these patients, they were not formed by XY donor-derived cells, donor BM cells were not associated with the stem cell (SC) niches assessed by telomapping technique, and engraftment percentages were very low with no correlation between time from transplant and engraftment efficiency, suggesting random terminal differentiation. In conclusion, XY donor-derived cells of a BM origin may be considered a limited exogenous source of transdifferentiated endometrial cells rather than a cyclic source of BM donor-derived stem cells.
Collapse
Affiliation(s)
- Irene Cervelló
- Fundación IVI-Instituto Universitario IVI-Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Claudia Gil-Sanchis
- Fundación IVI-Instituto Universitario IVI-Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Aymara Mas
- Fundación IVI-Instituto Universitario IVI-Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Amparo Faus
- Fundación IVI-Instituto Universitario IVI-Universidad de Valencia, INCLIVA, Valencia, Spain
| | - Jaime Sanz
- Servicio de Hematología, Hospital Universitario La Fe, Valencia, Spain
| | - Federico Moscardó
- Servicio de Hematología, Hospital Universitario La Fe, Valencia, Spain
| | - Gema Higueras
- Servicio de Ginecología, Hospital Universitario La Fe, Valencia, Spain
| | - Miguel Angel Sanz
- Servicio de Hematología, Hospital Universitario La Fe, Valencia, Spain
| | - Antonio Pellicer
- Servicio de Ginecología, Hospital Universitario La Fe, Valencia, Spain
| | - Carlos Simón
- Fundación IVI-Instituto Universitario IVI-Universidad de Valencia, INCLIVA, Valencia, Spain
- Stem Cell Bank, Centro de Investigación Príncipe Felipe, Valencia, Spain
- * E-mail:
| |
Collapse
|