1
|
Li J, Deng T, Zhu S, Xie P, Wang W, Zhou H, Xu C. The SDF-1/CXCR4 axis is involved in adipose-derived stem cell migration. Neurourol Urodyn 2024; 43:2279-2289. [PMID: 39149821 DOI: 10.1002/nau.25571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Intravenous injection of adipose-derived stem cells (ADSCs) can improve the urinary function of stress urinary incontinence (SUI) model rats and C-X-C chemokine receptor type 4 (CXCR4)-positive ADSCs are found in urethral tissues. The CXCR4 ligand stromal cell-derived factor-1 (SDF-1) is highly expressed in urinary incontinence model rats. In this study, we investigated the involvement of the SDF-1/CXCR4 axis in the homing of ADSCs. METHODS ADSCs were isolated from rats and purified. The levels of CXCR4 and CXCR7 were determined by western blot analysis and immunofluorescence assays following stimulation with SDF-1. Hypoxia conditioning was performed to treat the cells in vitro, following which the messenger RNA (mRNA) and protein level of SDF-1, CXCR4, and CXCR7 were estimated. RESULTS We found that CXCR4 and CXCR7 were expressed in ADSCs at passage zero (P0), P1, and P3, and the expression of both increased after SDF-1 stimulation. The level of expression of the mRNAs and proteins of SDF-1, CXCR4, and CXCR7 in ADSCs was higher after hypoxic conditioning. We then knocked down CXCR4 or CXCR7 using small interfering RNAs and found that the mRNA levels of CXCR4 and CXCR7 were considerably downregulated in the si-CXCR4/7-transfected cells. We also found that the SDF-1/CXCR4 axis was required for the migration of ADSCs. The phosphorylation levels of Janus kinase (JAK), protein kinase B (AKT), and extracellular regulated protein kinase significantly increased in SDF-1-stimulated ADSCs. However, the migration of ADSCs was suppressed when the corresponding specific inhibitors were used to block JAK and AKT signaling or silence CXCR4, whereas no significant change was observed in the migratory ability of ADSCs when the ERK pathway was blocked or CXCR7 was silenced. CONCLUSIONS The SDF-1/CXCR4 axis is involved in the migration of ADSCs and may play a role in the migrate of ADSCs in SUI.
Collapse
Affiliation(s)
- Jiang Li
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Tibin Deng
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Shaojie Zhu
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Pingbo Xie
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Wei Wang
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Hongqing Zhou
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| | - Chenxiang Xu
- The Second Ward of Urology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, China
| |
Collapse
|
2
|
Aleynik DY, Charykova IN, Rubtsova YP, Linkova DD, Farafontova EA, Egorikhina MN. Specific Features of the Functional Activity of Human Adipose Stromal Cells in the Structure of a Partial Skin-Equivalent. Int J Mol Sci 2024; 25:6290. [PMID: 38927998 PMCID: PMC11203524 DOI: 10.3390/ijms25126290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal adipose stromal cells (ASCs) are considered the most promising and accessible material for translational medicine. ASCs can be used independently or within the structure of scaffold-based constructs, as these not only ensure mechanical support, but can also optimize conditions for cell activity, as specific features of the scaffold structure have an impact on the vital activity of the cells. This manuscript presents a study of the secretion and accumulation that occur in a conditioned medium during the cultivation of human ASCs within the structure of such a partial skin-equivalent that is in contact with it. It is demonstrated that the ASCs retain their functional activity during cultivation both within this partial skin-equivalent structure and, separately, on plastic substrates: they proliferate and secrete various proteins that can then accumulate in the conditioned media. Our comparative study of changes in the conditioned media during cultivation of ASCs on plastic and within the partial skin-equivalent structure reveals the different dynamics of the release and accumulation of such secretory factors in the media under a variety of conditions of cell functioning. It is also demonstrated that the optimal markers for assessment of the ASCs' secretory functions in the studied partial skin-equivalent structure are the trophic factors VEGF-A, HGF, MCP, SDF-1α, IL-6 and IL-8. The results will help with the development of an algorithm for preclinical studies of this skin-equivalent in vitro and may be useful in studying various other complex constructs that include ASCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Marfa N. Egorikhina
- Federal State Budgetary Educational Institution of Higher Education, Privolzhsky Research Medical University of the Ministry of Health of the Russian Federation, 603005 Nizhny Novgorod, Russia; (D.Y.A.); (I.N.C.); (Y.P.R.); (D.D.L.); (E.A.F.)
| |
Collapse
|
3
|
Kolahi Azar H, Imanpour A, Rezaee H, Ezzatifar F, Zarei-Behjani Z, Rostami M, Azami M, Behestizadeh N, Rezaei N. Mesenchymal stromal cells and CAR-T cells in regenerative medicine: The homing procedure and their effective parameters. Eur J Haematol 2024; 112:153-173. [PMID: 37254607 DOI: 10.1111/ejh.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 06/01/2023]
Abstract
Mesenchymal stromal cells (MSCs) and chimeric antigen receptor (CAR)-T cells are two core elements in cell therapy procedures. MSCs have significant immunomodulatory effects that alleviate inflammation in the tissue regeneration process, while administration of specific chemokines and adhesive molecules would primarily facilitate CAR-T cell trafficking into solid tumors. Multiple parameters affect cell homing, including the recipient's age, the number of cell passages, proper cell culture, and the delivery method. In addition, several chemokines are involved in the tumor microenvironment, affecting the homing procedure. This review discusses parameters that improve the efficiency of cell homing and significant cell therapy challenges. Emerging comprehensive mechanistic strategies such as non-systemic and systemic homing that revealed a significant role in cell therapy remodeling were also reviewed. Finally, the primary implications for the development of combination therapies that incorporate both MSCs and CAR-T cells for cancer treatment were discussed.
Collapse
Affiliation(s)
- Hanieh Kolahi Azar
- Department of Pathology, Tabriz University of Medical Sciences, Tabriz, Iran
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Aylar Imanpour
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh Rezaee
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ezzatifar
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Molecular and Cell Biology Research Center, Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zeinab Zarei-Behjani
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, Advanced School of Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Rostami
- Division of Food Safety and Hygiene, Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Food Science and Nutrition Group (FSAN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mahmoud Azami
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Behestizadeh
- Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
4
|
Xiao Y, Xu RH, Dai Y. Nanoghosts: Harnessing Mesenchymal Stem Cell Membrane for Construction of Drug Delivery Platforms Via Optimized Biomimetics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304824. [PMID: 37653618 DOI: 10.1002/smll.202304824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/10/2023] [Indexed: 09/02/2023]
Abstract
Mesenchymal stem cells (MSCs) are becoming hotspots for application in disease therapies recently, combining with biomaterials and drug delivery system. A major advantage of MSCs applied in drug delivery system is that these cells enable specific targeting and releasing of cargos to the disease sites. However, the potential tumor tropic effects of MSCs raised concerns on biosafety. To solve this problem, there are emerging methods of isolating cell membranes and developing nanoformulations to perform drug delivery, which avoids concerns on biosafety without disturbing the membrane functions of specific polarizing and locating. These cargoes are so called "nanoghosts." This review article summarizes the current applications of nanoghosts, the promising potential of MSCs to be applied in membrane isolation and nanoghost construction, and possible approaches to develop better drug delivery system harnessing from MSC ghost cell membranes.
Collapse
Affiliation(s)
- Yuan Xiao
- Faculty of Health Sciences and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| | - Ren-He Xu
- Faculty of Health Sciences and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| | - Yunlu Dai
- Faculty of Health Sciences and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| |
Collapse
|
5
|
Perez-Estenaga I, Chevalier MT, Peña E, Abizanda G, Alsharabasy AM, Larequi E, Cilla M, Perez MM, Gurtubay J, Garcia-Yebenes Castro M, Prosper F, Pandit A, Pelacho B. A Multimodal Scaffold for SDF1 Delivery Improves Cardiac Function in a Rat Subacute Myocardial Infarct Model. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50638-50651. [PMID: 37566441 PMCID: PMC10636708 DOI: 10.1021/acsami.3c04245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Ischemic heart disease is one of the leading causes of death worldwide. The efficient delivery of therapeutic growth factors could counteract the adverse prognosis of post-myocardial infarction (post-MI). In this study, a collagen hydrogel that is able to load and appropriately deliver pro-angiogenic stromal cell-derived factor 1 (SDF1) was physically coupled with a compact collagen membrane in order to provide the suture strength required for surgical implantation. This bilayer collagen-on-collagen scaffold (bCS) showed the suitable physicochemical properties that are needed for efficient implantation, and the scaffold was able to deliver therapeutic growth factors after MI. In vitro collagen matrix biodegradation led to a sustained SDF1 release and a lack of cytotoxicity in the relevant cell cultures. In vivo intervention in a rat subacute MI model resulted in the full integration of the scaffold into the heart after implantation and biocompatibility with the tissue, with a prevalence of anti-inflammatory and pro-angiogenic macrophages, as well as evidence of revascularization and improved cardiac function after 60 days. Moreover, the beneficial effect of the released SDF1 on heart remodeling was confirmed by a significant reduction in cardiac tissue stiffness. Our findings demonstrate that this multimodal scaffold is a desirable matrix that can be used as a drug delivery system and a scaffolding material to promote functional recovery after MI.
Collapse
Affiliation(s)
- Iñigo Perez-Estenaga
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | - Merari Tumin Chevalier
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Estefania Peña
- Aragon
Institute of Engineering Research, University
of Zaragoza, Zaragoza 50009, Spain
- CIBER-BBN—Centro
de Investigación Biomédica en Red en Bioingeniería
Biomateriales y Nanomedicina, Zaragoza 50018, Spain
| | - Gloria Abizanda
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
| | - Amir M. Alsharabasy
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Eduardo Larequi
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | - Myriam Cilla
- Aragon
Institute of Engineering Research, University
of Zaragoza, Zaragoza 50009, Spain
- CIBER-BBN—Centro
de Investigación Biomédica en Red en Bioingeniería
Biomateriales y Nanomedicina, Zaragoza 50018, Spain
| | - Marta M. Perez
- Department
of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Zaragoza 50009, Spain
| | - Jon Gurtubay
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | | | - Felipe Prosper
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
- Department
of Cell Therapy and Hematology, Clínica
Universidad de Navarra, Pamplona 31008, Spain
- CIBERONC, Madrid 28029, Spain
| | - Abhay Pandit
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Beatriz Pelacho
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
| |
Collapse
|
6
|
Rivas IL, Soltero-Rivera M, Vapniarsky N, Arzi B. Stromal cell therapy in cats with feline chronic gingivostomatitis: current perspectives and future direction. J Feline Med Surg 2023; 25:1098612X231185395. [PMID: 37548494 PMCID: PMC10811994 DOI: 10.1177/1098612x231185395] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Feline chronic gingivostomatitis (FCGS) is a painful, immune-mediated, oral mucosal inflammatory disease in cats. The etiology of FCGS remains unclear, with evidence pointing potentially toward a viral cause. Full-mouth tooth extraction is the current standard of care, and cats that are non-responsive to extraction therapy may need lifelong medical management and, in some cases, euthanasia. Adipose-derived mesenchymal stromal cells (adMSCs) have been demonstrated to have advantages in the treatment and potentially the cure of non-responsive FCGS in cats. Therefore, adMSCs have attracted a series of ongoing clinical trials in the past decade. AdMSC therapy immediately after full-mouth tooth extraction was not explored, and we postulate that it may benefit the overall success rate of FCGS therapy. Here, we aim to summarize the current knowledge and impact of adMSCs for the therapeutic management of FCGS and to suggest a novel modified approach to further increase the efficacy of FCGS treatment in cats.
Collapse
Affiliation(s)
- Iris L Rivas
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Maria Soltero-Rivera
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Natalia Vapniarsky
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
- Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|
7
|
Yang MY, Chiu CD, Ke YC, Yang YC, Chang KB, Chen CM, Lee HT, Tang CL, Liu BS, Hung HS. Differentiation Induction of Mesenchymal Stem Cells by a Au Delivery Platform. Cells 2023; 12:1893. [PMID: 37508556 PMCID: PMC10378595 DOI: 10.3390/cells12141893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/11/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Au decorated with type I collagen (Col) was used as a core material to cross-link with stromal cell-derived factor 1α (SDF1α) in order to investigate biological performance. The Au-based nanoparticles were subjected to physicochemical determination using scanning electron microscopy (SEM), dynamic light scattering (DLS) and ultraviolet-visible (UV-Vis) and Fourier-transform infrared spectroscopy (FTIR). Mesenchymal stem cells (MSCs) were used to evaluate the biocompatibility of this nanoparticle using the MTT assay and measuring reactive oxygen species (ROS) production. Also, the biological effects of the SDF-1α-conjugated nanoparticles (Au-Col-SDF1α) were assessed and the mechanisms were explored. Furthermore, we investigated the cell differentiation-inducing potential of these conjugated nanoparticles on MSCs toward endothelial cells, neurons, osteoblasts and adipocytes. We then ultimately explored the process of cell entry and transportation of the nanoparticles. Using a mouse animal model and retro-orbital sinus injection, we traced in vivo biodistribution to determine the biosafety of the Au-Col-SDF1α nanoparticles. In summary, our results indicate that Au-Col is a promising drug delivery system; it can be used to carry SDF1α to improve MSC therapeutic efficiency.
Collapse
Affiliation(s)
- Meng-Yin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan
- College of Nursing, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Cheng-Di Chiu
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
- Spine Center, China Medical University Hospital, Taichung 404327, Taiwan
| | - Yi-Chun Ke
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan
| | - Yi-Chin Yang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Kai-Bo Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
- Department of Leisure Industry Management, National Chin-Yi University of Technology, Taichung 411030, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407219, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Chien-Lun Tang
- Department of Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407219, Taiwan
| | - Bai-Shuan Liu
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan
| | - Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan
- Translational Medicine Research, China Medical University Hospital, Taichung 404327, Taiwan
| |
Collapse
|
8
|
Go YY, Lee CM, Chae SW, Song JJ. Regenerative capacity of trophoblast stem cell-derived extracellular vesicles on mesenchymal stem cells. Biomater Res 2023; 27:62. [PMID: 37370189 DOI: 10.1186/s40824-023-00396-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Human mesenchymal stem cells (MSCs) are therapeutic for clinical applications because of their excellent immunomodulatory and multiple lineage differentiation abilities at tissue injury sites. However, insufficient number of cells and lack of regenerative properties during in vitro expansion still limit the clinical applicability of MSC therapies. Here, we demonstrated a preconditioning strategy with trophoblast stem cell-derived extracellular vesicles (TSC-EVs) to boost the proliferation and regenerative capacity of MSCs. METHODS We employed cell proliferation analyses such as CCK8 and BrdU assays to determine the proliferation-promoting role of TSC-EVs on MSCs. Osteogenic effects of TSC-EVs on MSCs were assessed by alkaline phosphatase (ALP) activity, calcium assays, and calvarial bone defect animal models. For skin regenerative effects, skin wound mice model was exploited to analyze wound-healing rate in this study, as well as immunofluorescence and histological staining evaluates. We also performed the small RNA profiling and RNA-sequencing analyzes to understand the cellular mechanism of TSC-EVs on MSCs. RESULTS TSC-EVs significantly promoted MSC proliferation under xeno-free conditions and facilitated the therapeutic effects of MSCs, including osteogenesis, anti-senescence, and wound healing. Transcriptomic analysis also provided evidence that specific microRNAs in TSC-EVs and differentially expressed genes (DEGs) in TSC-EV-treated MSCs showed the possibility of TSC-EVs triggering the regenerative abilities of MSCs with cytokine interaction. Hence, we found that NGF/Akt signaling mediated the regenerative effects of TSC-EVs on MSCs as a particular cellular signaling pathway. CONCLUSION The results of this study demonstrated the functional properties of TSC-EVs on MSCs for MSC-based therapeutic applications, suggesting that TSC-EVs may serve as a potential preconditioning source for MSC therapy in the clinical field of regenerative medicine.
Collapse
Affiliation(s)
- Yoon-Young Go
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea
| | - Chan-Mi Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Sung-Won Chae
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Guro Hospital, 80 Guro-Dong, Guro-Gu, Seoul, 08308, South Korea.
- Institute for Health Care Convergence Center, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
9
|
Wang M, Zhan H, Wang J, Song H, Sun J, Zhao G. Calcium silicate-stimulated adipose-derived stem cells promote angiogenesis and improve skin wound healing. Aging (Albany NY) 2023; 15:204760. [PMID: 37263631 DOI: 10.18632/aging.204760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023]
Abstract
Skin wound healing is a complicated process involving proliferation, inflammation, coagulation, and hemostasis, and scar tissue formation of wound repairing. Adipose-derived stem cells (ADSCs) have presented potential therapeutic effects in the non-healing and chronic wound. Calcium silicate (CS) ceramics have been identified as a new type of bioceramics for tissue construction and regeneration. Here, we aimed to explore the impact of CS on the regulation of ADSCs-mediated wound healing. Significantly, CS was able to dose-dependently enhance the proliferation of ADSCs. CS inhibited terminal deoxynucleotidyl transferase dUTP nick end labeling positive cells in the H2O2-treated ADSCs. Similarly, the Bcl-2 expression was elevated while Bax and cleaved caspase-3 expression were repressed by CS in the cells. CS could induce migration and reduce oxidative stress of ADSCs. Moreover, immunofluorescence analysis and Western blot analysis showed that CS could promote CXCR4 expression in ADSCs. Moreover, CS-stimulated ADSCs enhanced migration and angiogenic capacity of HUVEC. Importantly, CS-stimulated ADSCs improved wound healing in full-thickness skin defect mouse model. Thus, we conclude that CS improves ADSCs-attenuated wound healing in vivo and in vitro. Our finding presents novel insight in the scenario that CS regulates ADSCs and wound healing. CS may be applied as potential materials for the treatment of wound healing.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Orthopaedics, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong, China
| | - Hongyan Zhan
- Department of B-Ultrasound, The Fourth People’s Hospital of Jinan, Jinan, Shandong, China
| | - Jianhua Wang
- Department of Orthopaedics, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong, China
| | - Hua Song
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong, China
| | - Jianhua Sun
- Department of Orthopaedics, Tengzhou Central People’s Hospital, Tengzhou, Shandong, China
| | - Gang Zhao
- Department of Orthopaedics, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
10
|
Cheng WY, Yang MY, Yeh CA, Yang YC, Chang KB, Chen KY, Liu SY, Tang CL, Shen CC, Hung HS. Therapeutic Applications of Mesenchymal Stem Cell Loaded with Gold Nanoparticles for Regenerative Medicine. Pharmaceutics 2023; 15:1385. [PMID: 37242627 PMCID: PMC10222259 DOI: 10.3390/pharmaceutics15051385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/14/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
In the present study, the various concentrations of AuNP (1.25, 2.5, 5, 10 ppm) were prepared to investigate the biocompatibility, biological performances and cell uptake efficiency via Wharton's jelly mesenchymal stem cells and rat model. The pure AuNP, AuNP combined with Col (AuNP-Col) and FITC conjugated AuNP-Col (AuNP-Col-FITC) were characterized by Ultraviolet-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR) and Dynamic Light Scattering (DLS) assays. For in vitro examinations, we explored whether the Wharton's jelly MSCs had better viability, higher CXCR4 expression, greater migration distance and lower apoptotic-related proteins expression with AuNP 1.25 and 2.5 ppm treatments. Furthermore, we considered whether the treatments of 1.25 and 2.5 ppm AuNP could induce the CXCR4 knocked down Wharton's jelly MSCs to express CXCR4 and reduce the expression level of apoptotic proteins. We also treated the Wharton's jelly MSCs with AuNP-Col to investigate the intracellular uptake mechanisms. The evidence demonstrated the cells uptake AuNP-Col through clathrin-mediated endocytosis and the vacuolar-type H+-ATPase pathway with good stability inside the cells to avoid lysosomal degradation as well as better uptake efficiency. Additionally, the results from in vivo examinations elucidated the 2.5 ppm of AuNP attenuated foreign body responses and had better retention efficacy with tissue integrity in animal model. In conclusion, the evidence demonstrates that AuNP shows promise as a biosafe nanodrug delivery system for development of regenerative medicine coupled with Wharton's jelly MSCs.
Collapse
Affiliation(s)
- Wen-Yu Cheng
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
- Department of Physical Therapy, Hung Kuang University, Taichung 433304, Taiwan
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Meng-Yin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung 402202, Taiwan
| | - Chun-An Yeh
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (C.-A.Y.); (K.-B.C.)
| | - Yi-Chin Yang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
| | - Kai-Bo Chang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (C.-A.Y.); (K.-B.C.)
| | - Kai-Yuan Chen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
| | - Szu-Yuan Liu
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
| | - Chien-Lun Tang
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
| | - Chiung-Chyi Shen
- Department of Minimally Invasive Skull Base Neurosurgery, Neurological Institute, Taichung Veterans General Hospital, Taichung 407204, Taiwan; (M.-Y.Y.); (Y.-C.Y.); (K.-Y.C.); (S.-Y.L.); (C.-L.T.); (C.-C.S.)
| | - Huey-Shan Hung
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan; (C.-A.Y.); (K.-B.C.)
- Translational Medicine Research, China Medical University Hospital, Taichung 404327, Taiwan
| |
Collapse
|
11
|
Umar AK, Luckanagul JA, Zothantluanga JH, Sriwidodo S. Complexed Polymer Film-Forming Spray: An Optimal Delivery System for Secretome of Mesenchymal Stem Cell as Diabetic Wound Dressing? Pharmaceuticals (Basel) 2022; 15:867. [PMID: 35890165 PMCID: PMC9324405 DOI: 10.3390/ph15070867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes-related wounds have physiological factors that make healing more complicated. High sugar levels can increase microbial infection risk while limiting nutrition and oxygen transfer to the wound area. The secretome of mesenchymal stem cells has been widely known for its efficacy in regenerative therapy. However, applying the secretome directly to the wound can reduce its effectiveness. In this review, we examined the literature on synthesizing the combinations of carboxymethyl chitosan, hyaluronic acid, and collagen tripeptides, as well as the possibility of physicochemical properties enhancement of the hydrogel matrix, which could potentially be used as an optimal delivery system of stem cell's secretome for diabetic wound healing.
Collapse
Affiliation(s)
- Abd. Kakhar Umar
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Jittima Amie Luckanagul
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - James H. Zothantluanga
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India;
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
12
|
Angiogenic Effects and Crosstalk of Adipose-Derived Mesenchymal Stem/Stromal Cells and Their Extracellular Vesicles with Endothelial Cells. Int J Mol Sci 2021; 22:ijms221910890. [PMID: 34639228 PMCID: PMC8509224 DOI: 10.3390/ijms221910890] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/13/2022] Open
Abstract
Adipose-derived mesenchymal stem/stromal cells (ASCs) are an adult stem cell population able to self-renew and differentiate into numerous cell lineages. ASCs provide a promising future for therapeutic angiogenesis due to their ability to promote blood vessel formation. Specifically, their ability to differentiate into endothelial cells (ECs) and pericyte-like cells and to secrete angiogenesis-promoting growth factors and extracellular vesicles (EVs) makes them an ideal option in cell therapy and in regenerative medicine in conditions including tissue ischemia. In recent angiogenesis research, ASCs have often been co-cultured with an endothelial cell (EC) type in order to form mature vessel-like networks in specific culture conditions. In this review, we introduce co-culture systems and co-transplantation studies between ASCs and ECs. In co-cultures, the cells communicate via direct cell-cell contact or via paracrine signaling. Most often, ASCs are found in the perivascular niche lining the vessels, where they stabilize the vascular structures and express common pericyte surface proteins. In co-cultures, ASCs modulate endothelial cells and induce angiogenesis by promoting tube formation, partly via secretion of EVs. In vivo co-transplantation of ASCs and ECs showed improved formation of functional vessels over a single cell type transplantation. Adipose tissue as a cell source for both mesenchymal stem cells and ECs for co-transplantation serves as a prominent option for therapeutic angiogenesis and blood perfusion in vivo.
Collapse
|
13
|
Induction of the CD24 Surface Antigen in Primary Undifferentiated Human Adipose Progenitor Cells by the Hedgehog Signaling Pathway. Biologics 2021. [DOI: 10.3390/biologics1020008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the murine model system of adipogenesis, the CD24 cell surface protein represents a valuable marker to label undifferentiated adipose progenitor cells. Indeed, when injected into the residual fat pads of lipodystrophic mice, these CD24 positive cells reconstitute a normal white adipose tissue (WAT) depot. Unluckily, similar studies in humans are rare and incomplete. This is because it is impossible to obtain large numbers of primary CD24 positive human adipose stem cells (hASCs). This study shows that primary hASCs start to express the glycosylphosphatidylinositol (GPI)-anchored CD24 protein when cultured with a chemically defined medium supplemented with molecules that activate the Hedgehog (Hh) signaling pathway. Therefore, this in vitro system may help understand the biology and role in adipogenesis of the CD24-positive hASCs. The induced cells’ phenotype was studied by flow cytometry, Real-Time Quantitative Polymerase Chain Reaction (RT-qPCR) techniques, and their secretion profile. The results show that CD24 positive cells are early undifferentiated progenitors expressing molecules related to the angiogenic pathway.
Collapse
|
14
|
An T, Chen Y, Tu Y, Lin P. Mesenchymal Stromal Cell-Derived Extracellular Vesicles in the Treatment of Diabetic Foot Ulcers: Application and Challenges. Stem Cell Rev Rep 2021; 17:369-378. [PMID: 32772239 DOI: 10.1007/s12015-020-10014-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetic foot ischemia and ulcer (DFU) persists as a serious diabetes mellitus complication in spite of increased understanding of the pathophysiology and the cellular and molecular responses. Contributing to this pessimistic situation is the lack of effective treatments that are slow to heal the deep chronic wounds and microvascular obstruction. Mesenchymal stromal cells (MSCs) have been tested as a promising cell-based therapy for diabetes in vitro and in vivo, which is able to accelerate wound closure with increased epithelialization, granulation tissue formation and angiogenesis by differentiation into skin cells and paracrine pathways to repair injured cells. The secretomes of MSCs, including cytokines, growth factors, chemokines, and extracellular vesicles containing mRNA, proteins and microRNAs, have immunomodulatory and regenerative effects. This review will shed new light on the therapeutic potential of MSC-derived extracellular vesicles (MSC-EVs) for the treatment of diabetes-induced lower limb ischemia and ulcers. The identification of underlying mechanisms for MSC-EVs regulation on impaired diabetic wound healing might provide a new direction for MSC-centered treatment for diabetic lower limb ischemia and ulcers. Immunomodulatory and angiogenic effects of MSC-derived extracellular vesicles on diabetic foot ulcer.
Collapse
Affiliation(s)
- Tao An
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Yi Chen
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Yingchun Tu
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China
| | - Ping Lin
- Department of hand and foot surgery, Jinhua Hospital of Zhejiang University, Jinhua, People's Republic of China.
- Department of hand and foot surgery, Jinhua Central Hospital, 365 Renmin East Road, Jinhua, Zhejiang Province, People's Republic of China.
| |
Collapse
|
15
|
Luo W, Gong Y, Qiu F, Yuan Y, Jia W, Liu Z, Gao L. NGF nanoparticles enhance the potency of transplanted human umbilical cord mesenchymal stem cells for myocardial repair. Am J Physiol Heart Circ Physiol 2021; 320:H1959-H1974. [PMID: 33769916 DOI: 10.1152/ajpheart.00855.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this study, we investigated whether human umbilical cord mesenchymal stem cell (hUCMSC) fibrin patches loaded with nerve growth factor (NGF) poly(lactic-co-glycolic acid) (PLGA) nanoparticles could enhance the therapeutic potency of hUCMSCs for myocardial infarction (MI). In vitro, NGF significantly improved the proliferation of hUCMSCs and mitigated cytotoxicity and apoptosis under hypoxic injury. NGF also promoted the paracrine effects of hUCMSCs on angiogenesis and cardiomyocyte protection. The tyrosine kinase A (TrkA) and phosphoinositide 3-kinase (PI3K)-serine/threonine protein kinase (Akt) signaling pathways in hUCMSCs were involved in the NGF-induced protection. NGF PLGA nanoparticles continued to release NGF for at least 1 mo and also exerted a protective effect on hUCMSCs, the same with free NGF. In vivo, we treated MI mice with nothing (MI group), a cell-free fibrin patch with blank PLGA nanoparticles (MI + OP group), a cell-free fibrin patch with NGF nanoparticles (MI + NGF group), and hUCMSC fibrin patches with blank PLGA nanoparticles (MI + MSC group) or NGF PLGA nanoparticles (MSC + NGF group). Among these groups, the MSC + NGF group exhibited the best cardiac contractile function, the smallest infarct size, and the thickest ventricular wall. The application of NGF PLGA nanoparticles significantly improved the retention of transplanted hUCMSCs and enhanced their ability to reduce myocardial apoptosis and promote angiogenesis in the mouse heart after MI. These findings demonstrate the promising therapeutic potential of hUCMSC fibrin cardiac patches loaded with NGF PLGA nanoparticles.NEW & NOTEWORTHY NGF PLGA nanoparticles can exert a protective effect on hUCMSCs and promote the paracrine effects of hUCMSCs on angiogenesis and cardiomyocyte protection through TrkA-PI3K/Akt signaling pathway, the same with free NGF. The application of NGF PLGA nanoparticles in the hUCMSC fibrin cardiac patches can significantly improve the retention of transplanted hUCMSCs and enhance their ability to reduce myocardial apoptosis and promote angiogenesis in the mouse heart after MI.
Collapse
Affiliation(s)
- Wei Luo
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanshan Gong
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fan Qiu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Yuan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenwen Jia
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongmin Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular and Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ling Gao
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Shanghai Institute of Stem Cell Research and Clinical translation, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
16
|
Kükrek H, Aitzetmüller M, Vodiškar M, Moog P, Machens HG, Duscher D. Erythropoetin can partially restore cigarette smoke induced effects on Adipose derived Stem Cells. Clin Hemorheol Microcirc 2021; 77:27-36. [PMID: 32651309 DOI: 10.3233/ch-200852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Adipose derived Stem Cells (ASCs) have been proven to play a key role in tissue regeneration. However, exposure to large amounts of cigarette smoke can drastically diminish their function. Erythropoetin (EPO), can modulate cellular response to injury. Therefore, we investigated the ability of EPO to restore the regenerative function and differentiation capacity of ASCs. MATERIAL AND METHODS Human ASCs were isolated from abdominoplasty samples using standard isolation procedures. Cell identity was established by means of Fluorescence Activated Cell Scanning. Subsequently, isolated ASCs were cultivated with cigarette smoke extract both with and without EPO. Parameters investigated included cellular metabolic activity, adipogenic and osteogenic differentiation capacity, and in vitro wound closure capacity. For further enhancing wound closure, EPO was combined with Granulocyte Macrophage Colony Stimulating Factor (GM-CSF) or Stromal Derived Factor-1 alpha (SDF-1 a). RESULTS Cigarette smoke reduces adipogenic differentiation, the osteogenic differentiation capacity as well as the in vitro wound healing ability of human derived ASCs. EPO did not change metabolic activity of ASCs significantly. The addition of EPO could partially restore their function. The combination of EPO with GM-CSF or SDF-1 did not result in a synergistic effect regarding wound healing ability. CONCLUSION Exposure to cigarette smoke significantly reduced the regenerative potential of ASCs. Treatment of ASCs exposed to cigarette smoke with EPO has the potential to partially restore their function.
Collapse
Affiliation(s)
- Haydar Kükrek
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Matthias Aitzetmüller
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Mateja Vodiškar
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Philipp Moog
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hans-Günther Machens
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dominik Duscher
- Department for Plastic and Hand Surgery, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
17
|
Wang MY, Wang YX, Li-Ling J, Xie HQ. Adult Stem Cell Therapy for Premature Ovarian Failure: From Bench to Bedside. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:63-78. [PMID: 33427039 DOI: 10.1089/ten.teb.2020.0205] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Premature ovarian failure (POF) is a devastating condition for women of childbearing age with serious health consequences, including distress, infertility, osteoporosis, autoimmune disorders, ischemic heart disease, and increased mortality. In addition to the mainstay estrogen therapy, stem cell therapy has been tested as the result of rapid progress in cell biology and reprogramming research. We hereby provide a review for the latest research and issues related with stem cell-based therapy for POF, and provide a commentary on various methods for enhancing its effect. Large amount of animal studies have demonstrated an extensive benefit of stem cells for failed ovarian recovering. As shown by such studies, stem cell therapy can result in recovery of hormonal levels, follicular activation, ovarian angiogenesis, and functional restoration. Meanwhile, a study of molecular pathways revealed that the function of stem cells mainly depends on their paracrine actions, which can produce multiple factors for the promotion of ovarian angiogenesis and regulation of cellular functions. Nevertheless, studies using disease models also revealed certain drawbacks. Clinical trials have shown that menstrual cycle and even pregnancy may occur in POF patients following transplantation of stem cells, although the limitations, including inadequate number of cases and space for the improvement of transplantation methodology. Only with its safety and effect get substantial improvement through laboratory experiments and clinical trials, can stem cell therapy really bring benefits to more patients. Additionally, effective pretreatment and appropriate transplantation methods for stem cells are also required. Taken together, stem cell therapy has shown a great potential for the reversal of POF and is stepping from bench to bedside. Impact statement Premature ovarian failure (POF) is a devastating condition with serious clinical consequences. The purpose of this review was to summarize the current status of stem cell therapy for POF. Considering the diversity of cell types and functions, a rigorous review is required for the guidance for further research into this field. Meanwhile, the challenges and prospect for clinical application of stem cell treatment, methodological improvements, and innovations are addressed.
Collapse
Affiliation(s)
- Ming-Yao Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yi-Xuan Wang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jesse Li-Ling
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Hui-Qi Xie
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, China
| |
Collapse
|
18
|
Laiva AL, O'Brien FJ, Keogh MB. SDF-1α gene-activated collagen scaffold enhances provasculogenic response in a coculture of human endothelial cells with human adipose-derived stromal cells. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:26. [PMID: 33677751 PMCID: PMC7936958 DOI: 10.1007/s10856-021-06499-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 02/17/2021] [Indexed: 05/06/2023]
Abstract
Novel biomaterials can be used to provide a better environment for cross talk between vessel forming endothelial cells and wound healing instructor stem cells for tissue regeneration. This study seeks to investigate if a collagen scaffold containing a proangiogenic gene encoding for the chemokine stromal-derived factor-1 alpha (SDF-1α GAS) could be used to enhance functional responses in a coculture of human umbilical vein endothelial cells (HUVECs) and human adipose-derived stem/stromal cells (ADSCs). Functional responses were determined by (1) monitoring the amount of junctional adhesion molecule VE-cadherin released during 14 days culture, (2) expression of provasculogenic genes on the 14th day, and (3) the bioactivity of secreted factors on neurogenic human Schwann cells. When we compared our SDF-1α GAS with a gene-free scaffold, the results showed positive proangiogenic determination characterized by a transient yet controlled release of the VE-cadherin. On the 14th day, the coculture on the SDF-1α GAS showed enhanced maturation than its gene-free equivalent through the elevation of provasculogenic genes (SDF-1α-7.4-fold, CXCR4-1.5-fold, eNOS-1.5-fold). Furthermore, we also found that the coculture on SDF-1α GAS secretes bioactive factors that significantly (p < 0.01) enhanced human Schwann cells' clustering to develop toward Bünger band-like structures. Conclusively, this study reports that SDF-1α GAS could be used to produce a bioactive vascularized construct through the enhancement of the cooperative effects between endothelial cells and ADSCs.
Collapse
Affiliation(s)
- Ashang L Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Department of Biomedical Science, Royal College of Surgeons in Ireland, Adliya, Bahrain
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Michael B Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
- Department of Biomedical Science, Royal College of Surgeons in Ireland, Adliya, Bahrain.
| |
Collapse
|
19
|
Laiva AL, O’Brien FJ, Keogh MB. SDF-1α Gene-Activated Collagen Scaffold Restores Pro-Angiogenic Wound Healing Features in Human Diabetic Adipose-Derived Stem Cells. Biomedicines 2021; 9:biomedicines9020160. [PMID: 33562165 PMCID: PMC7914837 DOI: 10.3390/biomedicines9020160] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Non-healing diabetic foot ulcers (DFUs) can lead to leg amputation in diabetic patients. Autologous stem cell therapy holds some potential to solve this problem; however, diabetic stem cells are relatively dysfunctional and restrictive in their wound healing abilities. This study sought to explore if a novel collagen-chondroitin sulfate (coll-CS) scaffold, functionalized with polyplex nanoparticles carrying the gene encoding for stromal-derived factor-1 alpha (SDF-1α gene-activated scaffold), can enhance the regenerative functionality of human diabetic adipose-derived stem cells (ADSCs). We assessed the impact of the gene-activated scaffold on diabetic ADSCs by comparing their response against healthy ADSCs cultured on a gene-free scaffold over two weeks. Overall, we found that the gene-activated scaffold could restore the pro-angiogenic regenerative response in the human diabetic ADSCs similar to the healthy ADSCs on the gene-free scaffold. Gene and protein expression analysis revealed that the gene-activated scaffold induced the overexpression of SDF-1α in diabetic ADSCs and engaged the receptor CXCR7, causing downstream β-arrestin signaling, as effectively as the transfected healthy ADSCs. The transfected diabetic ADSCs also exhibited pro-wound healing features characterized by active matrix remodeling of the provisional fibronectin matrix and basement membrane protein collagen IV. The gene-activated scaffold also induced a controlled pro-healing response in the healthy ADSCs by disabling early developmental factors signaling while promoting the expression of tissue remodeling components. Conclusively, we show that the SDF-1α gene-activated scaffold can overcome the deficiencies associated with diabetic ADSCs, paving the way for autologous stem cell therapies combined with novel biomaterials to treat DFUs.
Collapse
Affiliation(s)
- Ashang L. Laiva
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland; (A.L.L.); (F.J.O.)
- Department of Biomedical Science, Royal College of Surgeons in Ireland, Adliya, P.O. Box 15503 Manama, Bahrain
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland; (A.L.L.); (F.J.O.)
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
| | - Michael B. Keogh
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, Dublin 2, Ireland; (A.L.L.); (F.J.O.)
- Department of Biomedical Science, Royal College of Surgeons in Ireland, Adliya, P.O. Box 15503 Manama, Bahrain
- Correspondence: ; Tel.: +973-17351450
| |
Collapse
|
20
|
Alexeev V, Olavarria J, Bonaldo P, Merlini L, Igoucheva O. Congenital muscular dystrophy-associated inflammatory chemokines provide axes for effective recruitment of therapeutic adult stem cell into muscles. Stem Cell Res Ther 2020; 11:463. [PMID: 33138863 PMCID: PMC7607684 DOI: 10.1186/s13287-020-01979-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/16/2020] [Indexed: 11/10/2022] Open
Abstract
Background Congenital muscular dystrophies (CMD) are a clinically and genetically heterogeneous group of neuromuscular disorders characterized by muscle weakness. The two most prevalent forms of CMD, collagen VI-related myopathies (COL6RM) and laminin α2 deficient CMD type 1A (MDC1A), are both caused by deficiency or dysfunction of extracellular matrix proteins. Previously, we showed that an intramuscular transplantation of human adipose-derived stem cells (ADSC) into the muscle of the Col6a1−/− mice results in efficient stem cell engraftment, migration, long-term survival, and continuous production of the collagen VI protein, suggesting the feasibility of the systemic cellular therapy for COL6RM. In order for this therapeutic approach to work however, stem cells must be efficiently targeted to the entire body musculature. Thus, the main goal of this study is to test whether muscle homing of systemically transplanted ADSC can be enhanced by employing muscle-specific chemotactic signals originating from CMD-affected muscle tissue. Methods Proteomic screens of chemotactic molecules were conducted in the skeletal muscles of COL6RM- and MDC1A-affected patients and CMD mouse models to define the inflammatory and immune activities, thus, providing potential markers of disease activity or treatment effect. Also using a pre-clinical animal model, recapitulating mild Ullrich congenital muscular dystrophy (UCMD), the therapeutic relevance of identified chemotactic pathways was investigated in vivo, providing a basis for future clinical investigations. Results Comprehensive proteomic screens evaluating relevant human and mouse skeletal muscle biopsies offered chemotactic axes to enhance directional migration of systemically transplanted cells into CMD-affected muscles, including CCL5-CCR1/3/5, CCL2-CCR2, CXCL1/2-CXCR1,2, and CXCL7-CXCR2. Also, the specific populations of ADSC selected with an affinity for the chemokines being released by damaged muscle showed efficient migration to injured site and presented their therapeutic effect. Conclusions Collectively, identified molecules provided insight into the mechanisms governing directional migration and intramuscular trafficking of systemically infused stem cells, thus, permitting broad and effective application of the therapeutic adult stem cells for CMD treatment.
Collapse
Affiliation(s)
- Vitali Alexeev
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB, Rm. 430, Philadelphia, PA, 19107, USA
| | - Jacquelyn Olavarria
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB, Rm. 430, Philadelphia, PA, 19107, USA
| | - Paolo Bonaldo
- Departments of Molecular Medicine, University of Padova, Padova, Italy
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Olga Igoucheva
- Department of Dermatology and Cutaneous Biology, Jefferson Medical College, Thomas Jefferson University, 233 South 10th Street, BLSB, Rm. 430, Philadelphia, PA, 19107, USA.
| |
Collapse
|
21
|
Koch C, Engele J. Functions of the CXCL12 Receptor ACKR3/CXCR7-What Has Been Perceived and What Has Been Overlooked. Mol Pharmacol 2020; 98:577-585. [PMID: 32883765 DOI: 10.1124/molpharm.120.000056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/18/2022] Open
Abstract
The CXCL12 system is central to the development of many organs and is further crucially engaged in pathophysiological processes underlying cancer, inflammation, and cardiovascular disorders. This disease-associated role presently focuses major interest on the two CXCL12 receptors, CXCR4 and atypical chemokine receptor 3 (ACKR3)/CXCR7, as promising therapeutic targets. Major obstacles in these ongoing efforts are confusing reports on the differential use of either ACKR3/CXCR7 and/or CXCR4 across various cells as well as on the specific function(s) of ACKR3/CXCR7. Although basically no doubts remain that CXCR4 represents a classic chemokine receptor, functions assigned to ACKR3/CXCR7 range from those of a strictly silent scavenger receptor eventually modulating CXCR4 signaling to an active and independent signaling receptor. In this review, we depict a thorough analysis of our present knowledge on different modes of organization and functions of the cellular CXCL12 system. We further highlight the potential role of ACKR3/CXCR7 as a "crosslinker" of different receptor systems. Finally, we discuss mechanisms with the potency to impinge on the cellular organization of the CXCL12 system and hence might represent additional future therapeutic targets. SIGNIFICANCE STATEMENT: Delineating the recognized functions of atypical chemokine receptor 3 and CXCR4 in CXCL12 signaling is central to the more detailed understanding of the role of the CXCL12 system in health and disease and will help to guide future research efforts.
Collapse
Affiliation(s)
- Christian Koch
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| | - Jürgen Engele
- Institute of Anatomy, University of Leipzig, Medical Faculty, Leipzig, Germany
| |
Collapse
|
22
|
Alt EU, Winnier G, Haenel A, Rothoerl R, Solakoglu O, Alt C, Schmitz C. Towards a Comprehensive Understanding of UA-ADRCs (Uncultured, Autologous, Fresh, Unmodified, Adipose Derived Regenerative Cells, Isolated at Point of Care) in Regenerative Medicine. Cells 2020; 9:E1097. [PMID: 32365488 PMCID: PMC7290808 DOI: 10.3390/cells9051097] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
It has become practically impossible to survey the literature on cells derived from adipose tissue for regenerative medicine. The aim of this paper is to provide a comprehensive and translational understanding of the potential of UA-ADRCs (uncultured, unmodified, fresh, autologous adipose derived regenerative cells isolated at the point of care) and its application in regenerative medicine. We provide profound basic and clinical evidence demonstrating that tissue regeneration with UA-ADRCs is safe and effective. ADRCs are neither 'fat stem cells' nor could they exclusively be isolated from adipose tissue. ADRCs contain the same adult stem cells ubiquitously present in the walls of blood vessels that are able to differentiate into cells of all three germ layers. Of note, the specific isolation procedure used has a significant impact on the number and viability of cells and hence on safety and efficacy of UA-ADRCs. Furthermore, there is no need to specifically isolate and separate stem cells from the initial mixture of progenitor and stem cells found in ADRCs. Most importantly, UA-ADRCs have the physiological capacity to adequately regenerate tissue without need for more than minimally manipulating, stimulating and/or (genetically) reprogramming the cells for a broad range of clinical applications. Tissue regeneration with UA-ADRCs fulfills the criteria of homologous use as defined by the regulatory authorities.
Collapse
Affiliation(s)
- Eckhard U. Alt
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, USA
- Sanford Health, University of South Dakota, Sioux Falls, SD 57104, USA
- University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Isar Klinikum Munich, 80331 Munich, Germany
- InGeneron, Inc., Houston, TX 77054, USA
| | | | - Alexander Haenel
- Heart and Vascular Institute, Department of Medicine, Tulane University Health Science Center, New Orleans, LA 70112, USA
- Department of Radiology and Nuclear Medicine, University Hospital Schleswig-Holstein, 23562 Lübeck, Germany
| | | | - Oender Solakoglu
- Dental Department of the University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Periodontology and Implant Dentistry, 22453 Hamburg, Germany
| | | | - Christoph Schmitz
- Institute of Anatomy, Faculty of Medicine, LMU Munich, 80331 Munich, Germany
| |
Collapse
|
23
|
Mazini L, Rochette L, Admou B, Amal S, Malka G. Hopes and Limits of Adipose-Derived Stem Cells (ADSCs) and Mesenchymal Stem Cells (MSCs) in Wound Healing. Int J Mol Sci 2020; 21:E1306. [PMID: 32075181 PMCID: PMC7072889 DOI: 10.3390/ijms21041306] [Citation(s) in RCA: 258] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
Adipose tissue derived stem cells (ADSCs) are mesenchymal stem cells identified within subcutaneous tissue at the base of the hair follicle (dermal papilla cells), in the dermal sheets (dermal sheet cells), in interfollicular dermis, and in the hypodermis tissue. These cells are expected to play a major role in regulating skin regeneration and aging-associated morphologic disgraces and structural deficits. ADSCs are known to proliferate and differentiate into skin cells to repair damaged or dead cells, but also act by an autocrine and paracrine pathway to activate cell regeneration and the healing process. During wound healing, ADSCs have a great ability in migration to be recruited rapidly into wounded sites added to their differentiation towards dermal fibroblasts (DF), endothelial cells, and keratinocytes. Additionally, ADSCs and DFs are the major sources of the extracellular matrix (ECM) proteins involved in maintaining skin structure and function. Their interactions with skin cells are involved in regulating skin homeostasis and during healing. The evidence suggests that their secretomes ensure: (i) The change in macrophages inflammatory phenotype implicated in the inflammatory phase, (ii) the formation of new blood vessels, thus promoting angiogenesis by increasing endothelial cell differentiation and cell migration, and (iii) the formation of granulation tissues, skin cells, and ECM production, whereby proliferation and remodeling phases occur. These characteristics would be beneficial to therapeutic strategies in wound healing and skin aging and have driven more insights in many clinical investigations. Additionally, it was recently presented as the tool key in the new free-cell therapy in regenerative medicine. Nevertheless, ADSCs fulfill the general accepted criteria for cell-based therapies, but still need further investigations into their efficiency, taking into consideration the host-environment and patient-associated factors.
Collapse
Affiliation(s)
- Loubna Mazini
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Centre interface Applications Médicales (CIAM), Université Mohammed VI Polytechnique, Ben-Guerir 43 150, Morocco;
| | - Luc Rochette
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France;
| | - Brahim Admou
- Laboratoire d’immunologie, Centre de Recherche Clinique, Faculté de Médecine et Pharmacie, Université Cadi Ayyad, Centre Hospitalier Universitaire, Marrakech 40 000, Morocco;
| | - Said Amal
- Service de dermatologie, Faculté de Médecine et Pharmacie, Université Cadi Ayyad, Centre hospitalier universitaire, Marrakech 40000, Morocco;
| | - Gabriel Malka
- Laboratoire Cellules Souches et Régénération Cellulaire et Tissulaire, Centre interface Applications Médicales (CIAM), Université Mohammed VI Polytechnique, Ben-Guerir 43 150, Morocco;
| |
Collapse
|
24
|
Manferdini C, Paolella F, Gabusi E, Cattini L, Rojewski M, Schrezenmeier H, Addimanda O, Meliconi R, Lisignoli G. Osteoarthritic Milieu Affects Adipose-Derived Mesenchymal Stromal Cells. J Orthop Res 2020; 38:336-347. [PMID: 31424111 PMCID: PMC7003792 DOI: 10.1002/jor.24446] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/07/2019] [Indexed: 02/04/2023]
Abstract
The objective of this study was to define the effects of osteoarthritic (OA) milieu on good manufactured practice-adipose-derived mesenchymal stromal cells (GMP-ASC) that are commonly utilized in cell therapies. Two different OA milieu: OA synovial fluid (SF) and OA-conditioned medium (CM) from synoviocytes were used to treat GMP-ASC both in normoxia or hypoxia. GMP-ASC were tested for cell migration, proliferation, cytokine receptors expression (CXCR1, CXCR2, CXCR3, CXCR4, CXCR7, CCR1, CCR2, CCR3, CCR5, IL6R), and cytokines (CXCL8/IL8, CXCL10/IP10, CXCL12/SDF-1, CCL2/MCP1, CCL3/MIP1α, CCL4/MIP1β, CCL5/RANTES, IL6) release. Healthy SF was used as controls. We demonstrated that GMP-ASC show an increase in proliferation, migration, and modulation of CXCR1, CXCR3, CCR1, and CCR5 receptors in hypoxic condition. Moreover, GMP-ASC migration increased 15-fold when treated either with OA-SF or OA-CM compared with healthy SF both in normoxia and hypoxia. GMP-ASC treated in both OA milieu showed an increase in CXCR3, CCR3, and IL6R and a decrease in CCR1 and CCR2 receptors. In OA-SF, we detected higher amount of CXCL10/IP10 than in OA-CM, while CCL2/MCP1 and CCL4/MIP1β were higher in OA-CM compared with OA-SF. CXCL10/IP10 was the only chemokine of the OA milieu, which was down-modulated after treatment with GMP-ASC. In conclusion, we demonstrated specific effects of OA milieu on both GMP-ASC proliferation, migration, and cytokine receptor expression that were strictly dependent on the inflammatory and hypoxic environment. The use of characterized OA milieu is crucial to define the therapeutic effect of GMP-ASC and indicates that CXCL10/IP10-CXCR3 axis is partially involved in the GMP-ASC effect on synovial macrophages. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 38:336-347, 2020.
Collapse
Affiliation(s)
- Cristina Manferdini
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Francesca Paolella
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Elena Gabusi
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Luca Cattini
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| | - Markus Rojewski
- Institut für TransfusionsmedizinUniversität UlmUlmGermany,Institut für Klinische Transfusionsmedizin und ImmungenetikDRK‐Blutspendedienst Baden‐Württemberg–Hessen & UniversitätsklinikumUlmGermany
| | - Hubert Schrezenmeier
- Institut für TransfusionsmedizinUniversität UlmUlmGermany,Institut für Klinische Transfusionsmedizin und ImmungenetikDRK‐Blutspendedienst Baden‐Württemberg–Hessen & UniversitätsklinikumUlmGermany
| | - Olga Addimanda
- IRCCS Istituto Ortopedico RizzoliSSD Medicina e ReumatologiaBolognaItaly
| | - Riccardo Meliconi
- IRCCS Istituto Ortopedico RizzoliSSD Medicina e ReumatologiaBolognaItaly,Dipartimento di Scienze Biomediche e neuromotorieUniversità degli studi di BolognaBolognaItaly
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico RizzoliSC Laboratorio di Immunoreumatologia e Rigenerazione TissutaleBolognaItaly
| |
Collapse
|
25
|
Improved therapeutics of modified mesenchymal stem cells: an update. J Transl Med 2020; 18:42. [PMID: 32000804 PMCID: PMC6993499 DOI: 10.1186/s12967-020-02234-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 01/22/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) have attracted intense interest due to their powerful intrinsic properties of self-regeneration, immunomodulation and multi-potency, as well as being readily available and easy to isolate and culture. Notwithstanding, MSC based therapy suffers reduced efficacy due to several challenges which include unfavorable microenvironmental factors in vitro and in vivo. Body In the quest to circumvent these challenges, several modification techniques have been applied to the naïve MSC to improve its inherent therapeutic properties. These modification approaches can be broadly divided into two groups to include genetic modification and preconditioning modification (using drugs, growth factors and other molecules). This field has witnessed great progress and continues to gather interest and novelty. We review these innovative approaches in not only maintaining, but also enhancing the inherent biological activities and therapeutics of MSCs with respect to migration, homing to target site, adhesion, survival and reduced premature senescence. We discuss the application of the improved modified MSC in some selected human diseases. Possible ways of yet better enhancing the therapeutic outcome and overcoming challenges of MSC modification in the future are also elaborated. Conclusion The importance of prosurvival and promigratory abilities of MSCs in their therapeutic applications can never be overemphasized. These abilities are maintained and even further enhanced via MSC modifications against the inhospitable microenvironment during culture and transplantation. This is a turning point in MSC-based therapy with promising preclinical studies and higher future prospect.
Collapse
|
26
|
Yaman S, Chintapula U, Rodriguez E, Ramachandramoorthy H, Nguyen KT. Cell-mediated and cell membrane-coated nanoparticles for drug delivery and cancer therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:879-911. [PMID: 33796822 PMCID: PMC8011581 DOI: 10.20517/cdr.2020.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Nanotechnology-based drug delivery platforms have been developed over the last two decades because of their favorable features in terms of improved drug bioavailability and stability. Despite recent advancement in nanotechnology platforms, this approach still falls short to meet the complexity of biological systems and diseases, such as avoiding systemic side effects, manipulating biological interactions and overcoming drug resistance, which hinders the therapeutic outcomes of the NP-based drug delivery systems. To address these issues, various strategies have been developed including the use of engineered cells and/or cell membrane-coated nanocarriers. Cell membrane receptor profiles and characteristics are vital in performing therapeutic functions, targeting, and homing of either engineered cells or cell membrane-coated nanocarriers to the sites of interest. In this context, we comprehensively discuss various cell- and cell membrane-based drug delivery approaches towards cancer therapy, the therapeutic potential of these strategies, and the limitations associated with engineered cells as drug carriers and cell membrane-associated drug nanocarriers. Finally, we review various cell types and cell membrane receptors for their potential in targeting, immunomodulation and overcoming drug resistance in cancer.
Collapse
Affiliation(s)
- Serkan Yaman
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Yaman S and Chintapula U contributed equally to this work
| | - Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Yaman S and Chintapula U contributed equally to this work
| | - Edgar Rodriguez
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Harish Ramachandramoorthy
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence Address: Dr. Kytai T. Nguyen, Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd ERB244, Arlington, TX 76010, USA. E-mail:
| |
Collapse
|
27
|
Zhong J, Li J, Wei J, Huang D, Huo L, Zhao C, Lin Y, Chen W, Wei Y. Plumbagin Restrains Hepatocellular Carcinoma Angiogenesis by Stromal Cell-Derived Factor (SDF-1)/CXCR4-CXCR7 Axis. Med Sci Monit 2019; 25:6110-6119. [PMID: 31415486 PMCID: PMC6707097 DOI: 10.12659/msm.915782] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/25/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Anti-angiogenic therapy has recently emerged as a highly promising therapeutic strategy for treating hepatocellular carcinoma (HCC). MATERIAL AND METHODS We assessed cellular proliferation, invasion, and activation of growth factors (VEGF and IL-8) with SDF-1 induced in the hepatocellular carcinoma cell line SMMC-7721, and this progression was limited by plumbagin (PL). The human umbilical vein endothelial cell line HUVEC was co-cultured with SDF-1-induced SMMC-7721, and the expressions of CXCR7, CXCR4, and PI3K/Akt pathways after PL treatment were detected by RT-PCR and Western blot analysis. RESULTS The treatment of the hepatoma cell line SMMC-7721 with SDF-1 resulted in enhanced secretion of the angiogenic factors, IL-8 and VEGF, and shows that these stimulatory effects are abolished by PL. The study further demonstrated that PL not only abolishes SDF-1-induced formation of endothelial tubes, but also inhibits expression of CXCR4 and CXCR7, and partially prevents activation of angiogenic signaling pathways. CONCLUSIONS The effect of PL on the SDF-1-CXCR4/CXCR7 axis has become an attractive target for inhibiting angiogenesis in hepatoma cells. Our results provide more evidence for the clinical application of PL as part of traditional Chinese medicine in modern cancer treatment.
Collapse
Affiliation(s)
- Jing Zhong
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Junxuan Li
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Jiexiao Wei
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Delun Huang
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Lini Huo
- Department of Organic Chemistry, Faculty of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Chuan Zhao
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Yuning Lin
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Wanjun Chen
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Yanfei Wei
- Department of Physiology, Faculty of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| |
Collapse
|
28
|
Ullah M, Liu DD, Thakor AS. Mesenchymal Stromal Cell Homing: Mechanisms and Strategies for Improvement. iScience 2019; 15:421-438. [PMID: 31121468 PMCID: PMC6529790 DOI: 10.1016/j.isci.2019.05.004] [Citation(s) in RCA: 314] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/30/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been widely investigated for their therapeutic potential in regenerative medicine, owing to their ability to home damaged tissue and serve as a reservoir of growth factors and regenerative molecules. As such, clinical applications of MSCs are reliant on these cells successfully migrating to the desired tissue following their administration. Unfortunately, MSC homing is inefficient, with only a small percentage of cells reaching the target tissue following systemic administration. This attrition represents a major bottleneck in realizing the full therapeutic potential of MSC-based therapies. Accordingly, a variety of strategies have been employed in the hope of improving this process. Here, we review the molecular mechanisms underlying MSC homing, based on a multistep model involving (1) initial tethering by selectins, (2) activation by cytokines, (3) arrest by integrins, (4) diapedesis or transmigration using matrix remodelers, and (5) extravascular migration toward chemokine gradients. We then review the various strategies that have been investigated for improving MSC homing, including genetic modification, cell surface engineering, in vitro priming of MSCs, and in particular, ultrasound techniques, which have recently gained significant interest. Contextualizing these strategies within the multistep homing model emphasizes that our ability to optimize this process hinges on our understanding of its molecular mechanisms. Moving forward, it is only with a combined effort of basic biology and translational work that the potential of MSC-based therapies can be realized.
Collapse
Affiliation(s)
- Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| | - Daniel D Liu
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, CA 94304, USA.
| |
Collapse
|
29
|
Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother 2018; 109:2022-2034. [PMID: 30551458 DOI: 10.1016/j.biopha.2018.11.068] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/17/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023] Open
Abstract
Ischemic diseases refer to a wide range of diseases caused by reduced blood flow and a subsequently deficient oxygen and nutrient supply. The pathogenesis of ischemia is multifaceted and primarily involves inflammation, oxidative stress and an apoptotic response. Over the last decade, mesenchymal stem cells (MSCs) have been widely studied as potential cell therapy agents for ischemic diseases due to their multiple favourable functions. However, the low homing and survival rates of transplanted cells have been concerns limiting for their clinical application. Recently, increasing studies have attempted to enhance the efficacy of MSCs by various strategies including genetic modification, pretreatment, combined application and biomaterial application. The purpose of this review is to summarize these creative strategies and the progress in basic and preclinical studies.
Collapse
|
30
|
Zhu H, Wang X, Han Y, Zhang W, Xin W, Zheng X, Zhang J. Icariin promotes the migration of bone marrow stromal cells via the SDF-1α/HIF-1α/CXCR4 pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:4023-4031. [PMID: 30538428 PMCID: PMC6254989 DOI: 10.2147/dddt.s179989] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Purpose In this study, a series of in vitro experiments were performed to investigate the molecular mechanisms underlying cell migration promoted by icariin (ICA) at low concentrations. Materials and methods Bone marrow stromal cells (BMSCs) were cultured with different concentrations of ICA to verify whether it can enhance the efficiency of BMSCs migration. Western blot was employed to measure the expression of hypoxia-inducible factor-1α (HIF-1α) and C-X-C chemokine receptor type 4 (CXCR4) at different time points in BMSCs treated with ICA. Subsequently, we evaluated the function of HIF-1α in the expression of CXCR4 and the migration of cells by transfecting plasmid HIF-1α small interfering RNA (siHIF-1α) into BMSCs model. Results Our data indicated that different concentrations of ICA (10, 1, and 0.1 µM) further enhanced the chemotactic capability of SDF-1α, and the most prominent cell migration stimulatory effect was observed with 1 µM ICA. Furthermore, ICA significantly enhanced the protein levels of CXCR4 and HIF-1α, and this effect was blocked by ICI 12,780 (estrogen receptor antagonis). Moreover, transfection of BMSCs with siHIF-1α reduced CXCR4 expression, suggesting that HIF-1α can regulate the migration of cells by influencing the expression of CXCR4. Conclusion ICA promoted BMSCs migration via the activation of HIF-1α and further regulated the expression of CXCR4, suggesting that ICA might have beneficial effects in stem cell therapy.
Collapse
Affiliation(s)
- Haiyan Zhu
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China, .,Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong Province, China, .,Department of Stomatology, Weihai Municipal Hospital, Weihai, Shandong Province, China
| | - Xuxia Wang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China, .,Department of Oral and Maxillofacial Surgery, School of Stomatology, Shandong University, Jinan, Shandong Province, China
| | - Yuanyuan Han
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China, .,Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong Province, China,
| | - Wenjuan Zhang
- Department of Stomatology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Wei Xin
- Department of Stomatology, Weihai Municipal Hospital, Weihai, Shandong Province, China
| | - Xiaotao Zheng
- Department of Stomatology, Weihai Municipal Hospital, Weihai, Shandong Province, China
| | - Jun Zhang
- Shandong Provincial Key Laboratory of Oral Tissue Regeneration, School of Stomatology, Shandong University, Jinan, Shandong Province, China, .,Department of Orthodontics, School of Stomatology, Shandong University, Jinan, Shandong Province, China,
| |
Collapse
|
31
|
Deconstructing Adipogenesis Induced by β3-Adrenergic Receptor Activation with Single-Cell Expression Profiling. Cell Metab 2018; 28:300-309.e4. [PMID: 29937373 PMCID: PMC6082711 DOI: 10.1016/j.cmet.2018.05.025] [Citation(s) in RCA: 208] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/05/2018] [Accepted: 05/25/2018] [Indexed: 11/22/2022]
Abstract
Recruitment of brown/beige adipocytes (BAs) in white adipose tissue (WAT) involves proliferation and differentiation of adipocyte stem cells (ASCs) in concert with close interactions with resident immune cells. To deconvolve stromal cell heterogeneity in a comprehensive and unbiased fashion, we performed single-cell RNA sequencing (scRNA-seq) of >33,000 stromal/vascular cells from epididymal WAT (eWAT) and inguinal WAT (iWAT) under control conditions and during β3-adrenergic receptor (ADRB3) activation. scRNA-seq identified distinct ASC subpopulations in eWAT and iWAT that appeared to be differentially poised to enter the adipogenic pathway. ADRB3 activation triggered the dramatic appearance of proliferating ASCs in eWAT, whose differentiation into BAs could be inferred from a single time point. scRNA-seq identified various immune cell types in eWAT, including a proliferating macrophage subpopulation that occupies adipogenic niches. These results demonstrate the power of scRNA-seq to deconstruct adipogenic niches and suggest novel functional interactions among resident stromal cell subpopulations.
Collapse
|
32
|
Ranganath SH. Bioengineered cellular and cell membrane-derived vehicles for actively targeted drug delivery: So near and yet so far. Adv Drug Deliv Rev 2018; 132:57-80. [PMID: 29935987 DOI: 10.1016/j.addr.2018.06.012] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/16/2022]
Abstract
Cellular carriers for drug delivery are attractive alternatives to synthetic nanoparticles owing to their innate homing/targeting abilities. Here, we review molecular interactions involved in the homing of Mesenchymal stem cells (MSCs) and other cell types to understand the process of designing and engineering highly efficient, actively targeting cellular vehicles. In addition, we comprehensively discuss various genetic and non-genetic strategies and propose futuristic approaches of engineering MSC homing using micro/nanotechnology and high throughput small molecule screening. Most of the targeting abilities of a cell come from its plasma membrane, thus, efforts to harness cell membranes as drug delivery vehicles are gaining importance and are highlighted here. We also recognize and report the lack of detailed characterization of cell membranes in terms of safety, structural integrity, targeting functionality, and drug transport. Finally, we provide insights on future development of bioengineered cellular and cell membrane-derived vesicles for successful clinical translation.
Collapse
Affiliation(s)
- Sudhir H Ranganath
- Bio-INvENT Lab, Department of Chemical Engineering, Siddaganga Institute of Technology, B.H. Road, Tumakuru, 572103, Karnataka, India.
| |
Collapse
|
33
|
Lee TJ, Shim MS, Yu T, Choi K, Kim DI, Lee SH, Bhang SH. Bioreducible Polymer Micelles Based on Acid-Degradable Poly(ethylene glycol)-poly(amino ketal) Enhance the Stromal Cell-Derived Factor-1α Gene Transfection Efficacy and Therapeutic Angiogenesis of Human Adipose-Derived Stem Cells. Int J Mol Sci 2018; 19:ijms19020529. [PMID: 29425184 PMCID: PMC5855751 DOI: 10.3390/ijms19020529] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 01/09/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) have the potential to treat ischemic diseases. In general, ADSCs facilitate angiogenesis by secreting various pro-angiogenic growth factors. However, transplanted ADSCs have a low therapeutic efficacy in ischemic tissues due to their poor engraftment and low viability. Stromal cell-derived factor-1α (SDF-1α) improves the survival rate of stem cells transplanted into ischemic regions. In this study, we developed acid-degradable poly(ethylene glycol)-poly(amino ketal) (PEG-PAK)-based micelles for efficient intracellular delivery of SDF-1α plasmid DNA. The SDF-1α gene was successfully delivered into human ADSCs (hADSCs) using PEG-PAK micelles. Transfection of SDF-1α increased SDF-1α, vascular endothelial growth factor, and basic fibroblast growth factor gene expression and decreased apoptotic activity in hADSCs cultured under hypoxic conditions in comparison with conventional gene transfection using polyethylenimine. SDF-1α-transfected hADSCs also showed significantly increased SDF-1α and VEGF expression together with reduced apoptotic activity at 4 weeks after transplantation into mouse ischemic hindlimbs. Consequently, these cells improved angiogenesis in ischemic hindlimb regions. These PEG-PAK micelles may lead to the development of a novel therapeutic modality for ischemic diseases based on an acid-degradable polymer specialized for gene delivery.
Collapse
Affiliation(s)
- Tae-Jin Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea.
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 406-772, Korea.
| | - Taekyung Yu
- Department of Chemical Engineering, College of Engineering, Kyung Hee University, Youngin 17104, Korea.
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
- Developmental, Regenerative, and Stem Cell Biology Program, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Science, CHA University, Seongnam 463-400, Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea.
| |
Collapse
|
34
|
Rashid CS, Lien YC, Bansal A, Jaeckle-Santos LJ, Li C, Won KJ, Simmons RA. Transcriptomic Analysis Reveals Novel Mechanisms Mediating Islet Dysfunction in the Intrauterine Growth-Restricted Rat. Endocrinology 2018; 159:1035-1049. [PMID: 29309562 PMCID: PMC5793792 DOI: 10.1210/en.2017-00888] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023]
Abstract
Intrauterine growth restriction (IUGR) increases the risk of type 2 diabetes developing in adulthood. In previous studies that used bilateral uterine artery ligation in a rat model of IUGR, age-associated decline in glucose homeostasis and islet function was revealed. To elucidate mechanisms contributing to IUGR pathogenesis, the islet transcriptome was sequenced from 2-week-old rats, when in vivo glucose tolerance is mildly impaired, and at 10 weeks of age, when rats are hyperglycemic and have reduced β-cell mass. RNA sequencing and functional annotation with Ingenuity Pathway Analysis revealed temporal changes in IUGR islets. For instance, gene expression involving amino acid metabolism was significantly reduced primarily at 2 weeks of age, but ion channel expression, specifically that involved in cell-volume regulation, was more disrupted in adult IUGR islets. Additionally, we observed alterations in the microenvironment of IUGR islets with extracellular matrix genes being significantly increased at 2 weeks of age and significantly decreased at 10 weeks. Specifically, hyaluronan synthase 2 expression and hyaluronan staining were increased in IUGR islets at 2 weeks of age (P < 0.05). Mesenchymal stromal cell-derived factors that have been shown to preserve islet allograft function, such as Anxa1, Cxcl12, and others, also were increased at 2 weeks and decreased in adult islets. Finally, comparisons of differentially expressed genes with those of type 2 diabetic human islets support a role for these pathways in human patients with diabetes. Together, these data point to new mechanisms in the pathogenesis of IUGR-mediated islet dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Cetewayo S. Rashid
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Division of Neonatology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Yu-Chin Lien
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Amita Bansal
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Division of Neonatology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Lane J. Jaeckle-Santos
- Division of Neonatology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Changhong Li
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
- Institute for Diabetes, Obesity, and Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kyoung-Jae Won
- Institute for Diabetes, Obesity, and Metabolism, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Department of Genetics, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Rebecca A. Simmons
- Center for Research on Reproduction and Women’s Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
- Division of Neonatology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| |
Collapse
|
35
|
Piovesana R, Melfi S, Fiore M, Magnaghi V, Tata AM. M2 muscarinic receptor activation inhibits cell proliferation and migration of rat adipose-mesenchymal stem cells. J Cell Physiol 2018; 233:5348-5360. [PMID: 29227527 DOI: 10.1002/jcp.26350] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs), also known as stromal mesenchymal stem cells, are multipotent cells, which can be found in many tissues and organs as bone marrow, adipose tissue and other tissues. In particular MSCs derived from Adipose tissue (ADSCs) are the most frequently used in regenerative medicine because they are easy to source, rapidly expandable in culture and excellent differentiation potential into adipocytes, chondrocytes, and other cell types. Acetylcholine (ACh), the most important neurotransmitter in Central nervous system (CNS) and peripheral nervous system (PNS), plays important roles also in non-neural tissue, but its functions in MSCs are still not investigated. Although MSCs express muscarinic receptor subtypes, their role is completely unknown. In the present work muscarinic cholinergic effects were characterized in rat ADSCs. Analysis by RT-PCR demonstrates that ADSCs express M1-M4 muscarinic receptor subtypes, whereas M2 is one of the most expressed subtype. For this reason, our attention was focused on M2 subtype. By using the selective M2 against Arecaidine Propargyl Ester (APE) we performed cell proliferation and migration assays demonstrating that APE causes cell growth and migration inhibition without affecting cell survival. Our results indicate that ACh via M2 receptors, may contribute to the maintaining of the ADSCs quiescent status. These data are the first evidence that ACh, via muscarinic receptors, might contribute to control ADSCs physiology.
Collapse
Affiliation(s)
- Roberta Piovesana
- Department of Biology and Biotechnologies "Charles Darwin,", Sapienza University of Rome, Rome, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Simona Melfi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Mario Fiore
- IBPM, Institute of Molecular Biology and Pathology, CNR, Rome, Italy
| | - Valerio Magnaghi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Ada Maria Tata
- Department of Biology and Biotechnologies "Charles Darwin,", Sapienza University of Rome, Rome, Italy.,Center of Neurobiology "Daniel Bovet,", "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
36
|
Qian T, Liu Y, Dong Y, Zhang L, Dong Y, Sun Y, Sun D. CXCR7 regulates breast tumor metastasis and angiogenesis in vivo and in vitro. Mol Med Rep 2017; 17:3633-3639. [PMID: 29257351 PMCID: PMC5802168 DOI: 10.3892/mmr.2017.8286] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/03/2017] [Indexed: 12/16/2022] Open
Abstract
The chemokine receptor CXCR7 is regarded as a scavenger receptor for CXCL12, and induces numerous key steps in tumor growth and metastasis. However, the exact molecular mechanism of CXCR7 regulation in breast tumor angiogenesis remains unknown. In the present study, the function of CXCR7 in breast tumors was investigated in vitro and in vivo. The breast cancer MDA-MB-231 cell line was used. Pharmacological inhibition of CXCR7 by CCX771 reduced breast tumor invasion, adhesion and metastasis. Furthermore, CXCR7 was essential for the tube formation of HUVECs in vitro, and for blood vessel formation in a Matrigel plug assay in vivo. In addition, vascular endothelial growth factor expression was also decreased in CCX771-treated MDA-MB-231 cells, indicating that CCX771 regulates tumor angiogenesis. The present results indicated that CXCR7 regulated breast cancer metastasis at multiple stages; additional understanding of CXCR7 in tumor environments may develop anti-metastatic therapy.
Collapse
Affiliation(s)
- Tingting Qian
- Laboratory Animal Centre, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yancheng Liu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, P.R. China
| | - Yan Dong
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, Anhui 239000, P.R. China
| | - Lei Zhang
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, Anhui 239000, P.R. China
| | - Yining Dong
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, Anhui 239000, P.R. China
| | - Yanhui Sun
- College of Biology and Food Engineering, Chuzhou University, Chuzhou, Anhui 239000, P.R. China
| | - Dongmei Sun
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, P.R. China
| |
Collapse
|
37
|
Zeng Y, Wang X, Yin B, Xia G, Shen Z, Gu W, Wu M. Role of the stromal cell derived factor-1/CXC chemokine receptor 4 axis in the invasion and metastasis of lung cancer and mechanism. J Thorac Dis 2017; 9:4947-4959. [PMID: 29312699 DOI: 10.21037/jtd.2017.10.138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Lung cancer is the most common tumor, and has the highest incidence and mortality rates among all malignant tumors. Since stromal cell derived factor-1 (SDF-1) and CXC chemokine receptor 4 (CXCR4) are specific to binding sites, they are more important than other members of the families for tumor invasion and metastasis. We herein aimed to investigate the role of the axis of chemokine SDF-1 and its receptor CXCR4 in the invasion and metastasis of lung cancer. Methods Sixty clinical non-small cell lung cancer (NSCLC) tissue samples were collected. The CXCR4 expressions in cancer, paracancerous and normal lung tissues were detected by immunocytochemical assay and PCR. Cells with CXCR4 overexpression (CXCR4-A549) were constructed. After induction with SDF-1, CXCR4-A549 and A549 cells were subjected to in vitro chemotaxis and invasion assays. Their proliferation and apoptosis were detected by flow cytometry. The activities of phosphoinositide 3-kinase/protein kinase B (AKT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK)-related signaling pathways were detected by Western blot. The downstream signaling molecules that may be activated by SDF-1/CXCR4 were analyzed. The expressions of vascular endothelial growth factor-C and matrix metalloproteinase-2 were detected by Western blot and PCR. A mouse model was established by subcutaneous inoculation of lung cancer cells. The effects of up-regulated CXCR4 expression on the migration of lung cancer cells in vitro and their tumorigenesis and metastasis in vivo were assessed. Results There was no expression in normal or paracancerous tissues. The expression of CXCR4 mRNA in lung cancer tissues was 83.3% (50/60). The expressions of CXCR4 in lung squamous cell carcinoma and adenocarcinoma were similar (P>0.05). The expression of CXCR4 was 76.9% (10/13) in highly differentiated carcinoma, 82.1% (23/28) in moderately differentiated carcinoma and 84.2% (16/19) in lowly differentiated carcinoma (P>0.05). The expression of CXCR4 was 72.7% (8/11) in TNM stage I patients, 83.9% (26/31) in stage II patients, and 88.9% (16/18) in stage III patients, with significant correlations. After up-regulation of CXCR4, the invasion ability of CXCR4-A549 cells was increased 1.62-fold (P<0.05). ERK and AKT were significantly phosphorylated 30 min after SDF-1 treatment. The tumorigenic rates of six mice inoculated with CXCR4-A549 and A549 cells were both 100%, with the average tumor weights of (4.37±0.96 g) and (3.24±1.16 g) respectively (P<0.05). In the CXCR4-A549 group, metastatic tumors clearly formed in the lungs of 6 mice, but only 2 mice in the A549 group had tumor cell invasion. Conclusions SDF-1/CXCR4 played a key role in the invasion and metastasis of lung cancer. The interaction between SDF-1α and CXCR4 activated a series of downstream molecules by activating ERK and AKT.
Collapse
Affiliation(s)
- Yun Zeng
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China
| | - Xinwei Wang
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China
| | - Bijian Yin
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China
| | - Guohao Xia
- Department of Medical Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China
| | - Zhengjie Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wenzhe Gu
- Department of Otorhinolaryngology, Zhangjiagang Hospital of Traditional Chinese Medicine, Zhangjiagang 215600, China
| | - Mianhua Wu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
38
|
Johnson V, Webb T, Norman A, Coy J, Kurihara J, Regan D, Dow S. Activated Mesenchymal Stem Cells Interact with Antibiotics and Host Innate Immune Responses to Control Chronic Bacterial Infections. Sci Rep 2017; 7:9575. [PMID: 28851894 PMCID: PMC5575141 DOI: 10.1038/s41598-017-08311-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic bacterial infections associated with biofilm formation are often difficult to resolve without extended courses of antibiotic therapy. Mesenchymal stem cells (MSC) exert antibacterial activity in vitro and in acute bacterial infection models, but their activity in chronic infection with biofilm models has not been previously investigated. Therefore, we studied the effects of MSC administration in mouse and dog models of chronic infections associated with biofilms. Mice with chronic Staphylococcus aureus implant infections were treated by i.v. administration of activated or non-activated MSC, with or without antibiotic therapy. The most effective treatment protocol was identified as activated MSC co-administered with antibiotic therapy. Activated MSC were found to accumulate in the wound margins several days after i.v. administration. Macrophages in infected tissues assumed an M2 phenotype, compared to untreated infections which contained predominately M1 macrophages. Bacterial killing by MSC was found to be mediated in part by secretion of cathelicidin and was significantly increased by antibiotics. Studies in pet dogs with spontaneous chronic multi drug-resistant wound infections demonstrated clearance of bacteria and wound healing following repeated i.v. administration of activated allogeneic canine MSC. Thus, systemic therapy with activated MSC may be an effective new, non-antimicrobial approach to treatment of chronic, drug-resistant infections.
Collapse
Affiliation(s)
- Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Tracy Webb
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Annalis Norman
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jonathan Coy
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jade Kurihara
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Daniel Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA.
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW In the process of bone fracture healing, inflammation is thought to be an essential process that precedes bone formation and remodeling. We review recent studies on bone fracture healing from an osteoimmunological point of view. RECENT FINDINGS Based on previous observations that many types of immune cells infiltrate into the bone injury site and release a variety of molecules, recent studies have addressed the roles of specific immune cell subsets. Macrophages and interleukin (IL)-17-producing γδ T cells enhance bone healing, whereas CD8+ T cells impair bone repair. Additionally, IL-10-producing B cells may contribute to bone healing by suppressing excessive and/or prolonged inflammation. Although the involvement of other cells and molecules has been suggested, the precise underlying mechanisms remain elusive. Accumulating evidence has begun to reveal the deeper picture of bone fracture healing. Further studies are required for the development of novel therapeutic strategies for bone fracture.
Collapse
Affiliation(s)
- Takehito Ono
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
40
|
Mesenchymal Stem Cell Administration in Patients with Chronic Obstructive Pulmonary Disease: State of the Science. Stem Cells Int 2017; 2017:8916570. [PMID: 28303154 PMCID: PMC5337878 DOI: 10.1155/2017/8916570] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/22/2017] [Indexed: 01/10/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have chronic, irreversible airway inflammation; currently, there is no effective or curative treatment and the main goals of COPD management are to mitigate symptoms and improve patients' quality of life. Stem cell based therapy offers a promising therapeutic approach that has shown potential in diverse degenerative lung diseases. Preclinical studies have demonstrated encouraging outcomes of mesenchymal stem/stromal cells (MSCs) therapy for lung disorders including emphysema, bronchopulmonary dysplasia, fibrosis, and acute respiratory distress syndrome. This review summarizes available data on 15 studies currently registered by the ClinicalTrials.gov repository, which used different stem cell therapy protocols for COPD; these included bone marrow mononuclear cells (BMMCs), bone marrow-derived MSCs, adipose-derived stem/stromal cells (ADSCs), and adipose-derived MSCs. Published results of three trials indicate that administering BMMCs or MSCs in the setting of degenerative lung disease is safe and may improve patients' condition and quality of life; however, larger-scale studies are needed to evaluate efficacy. Results of another completed trial (NCT01872624) are not yet published, and eleven other studies are ongoing; these include MSCs therapy in emphysema, several studies of ADSCs in COPD, another in idiopathic pulmonary fibrosis, and plerixafor mobilization of CD117 stem cells to peripheral blood.
Collapse
|
41
|
Tang X, Li X, Li Z, Liu Y, Yao L, Song S, Yang H, Li C. Downregulation of CXCR7 inhibits proliferative capacity and stem cell-like properties in breast cancer stem cells. Tumour Biol 2016; 37:13425-13433. [PMID: 27460092 DOI: 10.1007/s13277-016-5180-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 07/12/2016] [Indexed: 01/07/2023] Open
Abstract
Breast cancer stem cells (bCSCs) are considered an obstacle in breast cancer therapy because they exhibit long-term proliferative potential, phenotypic plasticity and high resistance to the current therapeutics. CXC chemokine receptor type 7 (CXCR7), which provides a growth advantage to breast cancer cells, has recently been demonstrated to play an important role in the maintenance of stem cell-like properties in the CSCs of glioblastoma and lung cancer, yet its role in bCSCs remains elusive. In this study, CD44+/CD24low bCSC-enriched cells (bCSCs for short) were isolated from MCF-7 cells, and CXCR7 was stably knocked down in bCSCs via lentivirus-mediated transduction with CXCR7 short hairpin RNA (shRNA). Knockdown of CXCR7 in bCSCs decreased the proportion of CD44+/CD24low cells, and markedly reduced the clonogenicity of the cells. Moreover, silencing of CXCR7 downregulated the expression of stem cell markers, such as aldehyde dehydrogenase 1 (ALDH1), Oct4, and Nanog. In addition, CXCR7 silencing in bCSCs suppressed cell proliferation and G1/S transition in vitro, and delayed tumor growth in vivo in a xenograft mouse model. In situ immunohistochemical analysis revealed a reduction in Ki-67 expression and enhanced apoptosis in the xenograft tumors as a result of CXCR7 silencing. Furthermore, combined treatment with CXCR7 silencing and epirubicin displayed an outstanding anti-tumor effect compared with either single treatment. Our study demonstrates that CXCR7 plays a critical role in the maintenance of stem cell-like properties and promotion of growth in bCSCs, and suggests that CXCR7 may be a candidate target for bCSCs in breast cancer therapy.
Collapse
Affiliation(s)
- Xin Tang
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China
| | - Xiang Li
- Department of Medical Ultrasonics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zitao Li
- Department of Orthopaedic Surgery, Mudanjiang Forestry Central Hospital, Mudanjiang, Heilongjiang, 157000, China
| | - Yunshuang Liu
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China
| | - Lihong Yao
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China
| | - Shuang Song
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China
| | - Hongyan Yang
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China
| | - Caijuan Li
- Department of Medical Ultrasonics, Hongqi Hospital of Mudanjiang Medical University, 5 Tongxiang Road, Mudanjiang, Heilongjiang, 157011, China.
| |
Collapse
|
42
|
Li Q, Guo Y, Chen F, Liu J, Jin P. Stromal cell-derived factor-1 promotes human adipose tissue-derived stem cell survival and chronic wound healing. Exp Ther Med 2016; 12:45-50. [PMID: 27347016 PMCID: PMC4906949 DOI: 10.3892/etm.2016.3309] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 02/04/2015] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue-derived stem cells (ADSCs) hold great potential for the stem cell-based therapy of cutaneous wound healing. Stromal cell-derived factor-1 (SDF-1) activates CXC chemokine receptor (CXCR)4+ and CXCR7+ cells and plays an important role in wound healing. Increasing evidence suggests a critical role for SDF-1 in cell apoptosis and the survival of mesenchymal stem cells. However, the function of SDF-1 in the apoptosis and wound healing ability of ADSCs is not well understood. The aim of this study was to analyze the effect of SDF-1 on the apoptosis and therapeutic effect of ADSCs in cutaneous chronic wounds in vitro and in vivos. By flow cytometric analysis, it was found that hypoxia and serum free promoted the apoptosis of ADSCs. When pretreated with SDF-1, the apoptosis of ADSCs induced by hypoxia and serum depletion was partly recovered. Furthermore, in vivo experiments established that the post-implantation cell survival and chronic wound healing ability of ADSCs were increased following pretreatment with SDF-1 in a diabetic mouse model of chronic wound healing. To explore the potential mechanism underlying the effect of SDF-1 on ADSC apoptosis, western blot analysis was employed and the results indicate that SDF-1 may protect against cell apoptosis in hypoxic and serum-free conditions through activation of the caspase signaling pathway in ADSCs. This study provides evidence that SDF-1 pretreatment can increase the therapeutic effect of ADSCs in cutaneous chronic wounds in vitro and in vivo.
Collapse
Affiliation(s)
- Qiang Li
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China; Department of Ear Reconstruction, Plastic Surgery Hospital, Chinese Academy of Medical Sciences, Beijing 100144, P.R. China
| | - Yanping Guo
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Feifei Chen
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Jing Liu
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| | - Peisheng Jin
- Plastic Surgery Department, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
43
|
Genetic Engineering of Mesenchymal Stem Cells to Induce Their Migration and Survival. Stem Cells Int 2016; 2016:4956063. [PMID: 27242906 PMCID: PMC4868914 DOI: 10.1155/2016/4956063] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/22/2016] [Accepted: 03/14/2016] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are very attractive for regenerative medicine due to their relatively easy derivation and broad range of differentiation capabilities, either naturally or induced through cell engineering. However, efficient methods of delivery to diseased tissues and the long-term survival of grafted cells still need improvement. Here, we review genetic engineering approaches designed to enhance the migratory capacities of MSCs, as well as extend their survival after transplantation by the modulation of prosurvival approaches, including prevention of senescence and apoptosis. We highlight some of the latest examples that explore these pivotal points, which have great relevance in cell-based therapies.
Collapse
|
44
|
Herzmann N, Salamon A, Fiedler T, Peters K. Analysis of migration rate and chemotaxis of human adipose-derived mesenchymal stem cells in response to LPS and LTA in vitro. Exp Cell Res 2016; 342:95-103. [PMID: 26997527 DOI: 10.1016/j.yexcr.2016.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/17/2016] [Accepted: 03/16/2016] [Indexed: 11/18/2022]
Abstract
Mesenchymal stem cells (MSC) are able to stimulate the regeneration of injured tissue. Since bacterial infections are common complications in wound healing, bacterial pathogens and their components come into direct contact with MSC. The interaction with bacterial structures influences the proliferation, differentiation and migratory activity of the MSC, which might be of relevance during regeneration. Studies on MSC migration in response to bacterial components have shown different results depending on the cell type. Here, we analyzed the migration rate and chemotaxis of human adipose-derived MSC (adMSC) in response to the basic cell-wall components lipopolysaccharide (LPS) of Gram-negative bacteria and lipoteichoic acid (LTA) of Gram-positive bacteria in vitro. To this end, we used transwell and scratch assays, as well as a specific chemotaxis assay combined with live-cell imaging. We found no significant influence of LPS or LTA on the migration rate of adMSC in transwell or scratch assays. Furthermore, in the µ-slide chemotaxis assay, the stimulation with LPS did not exert any chemotactic effect on adMSC.
Collapse
Affiliation(s)
- Nicole Herzmann
- Department of Cell Biology, University Medicine Rostock, Schillingallee 69, D-18057 Rostock, Germany
| | - Achim Salamon
- Department of Cell Biology, University Medicine Rostock, Schillingallee 69, D-18057 Rostock, Germany
| | - Tomas Fiedler
- Institute for Medical Microbiology, Virology and Hygiene, University Medicine Rostock, Schillingallee 70, D-18057 Rostock, Germany
| | - Kirsten Peters
- Department of Cell Biology, University Medicine Rostock, Schillingallee 69, D-18057 Rostock, Germany.
| |
Collapse
|
45
|
Fotia C, Massa A, Boriani F, Baldini N, Granchi D. Prolonged exposure to hypoxic milieu improves the osteogenic potential of adipose derived stem cells. J Cell Biochem 2016; 116:1442-53. [PMID: 25648991 DOI: 10.1002/jcb.25106] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/27/2015] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSC) have been widely used in orthopedics for several applications. Conventionally, MSC are maintained under 21% O2 which does not reflect the real O2 tension in vivo. Recently, it was reported that different O2 conditions can give different cellular responses. Here, we investigated whether prolonged exposure to hypoxia affects the osteogenic differentiation of adipose-derived stem cells (ASC). ASC from six individuals were cultured under "low" (2-3%) or "air" (21%) oxygen tensions, either without or with osteogenic stimuli. The effect of the O2 tension was evaluated on cell proliferation, surface antigens, stemness and bone-related genes expression, alkaline phosphatase activity (ALP), mineralization activity, and release of osteogenic growth factors. Without differentiating stimuli, hypoxia favored ASC proliferation, reduced the number of CD184+ and CD34+ cells, and preserved the expression of NANOG and SOX2. The combination of hypoxia and osteogenic medium induced a high proliferation rate, a rapid and more pronounced mineralization activity, a higher expression of genes related to the MSC differentiation, a higher release of mitogenic growth factors (bFGF, PDGF-BB), and the decrease in TGF-β secretion, an inhibitor of the early stage of the osteoblast differentiation. We demonstrated that hypoxia acts dually, favoring ASC proliferation and the maintenance of the stemness in the absence of osteogenic stimuli, but inducing the differentiation in a bone-like microenvironment. In conclusion, prolonged cell culture in hypoxic microenvironment represents a proper method to modulate the stem cell function that may be used in several applications, for example, studies on bone pathophysiology or bone-tissue engineering.
Collapse
Affiliation(s)
- Caterina Fotia
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Annamaria Massa
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Filippo Boriani
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Nicola Baldini
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
46
|
Tobita M, Tajima S, Mizuno H. Adipose tissue-derived mesenchymal stem cells and platelet-rich plasma: stem cell transplantation methods that enhance stemness. Stem Cell Res Ther 2015; 6:215. [PMID: 26541973 PMCID: PMC4635588 DOI: 10.1186/s13287-015-0217-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Because of their ease of isolation and relative abundance, adipose-derived mesenchymal stem cells (ASCs) are a particularly attractive autologous cell source for various therapeutic purposes. ASCs retain a high proliferation capacity in vitro and have the ability to undergo extensive differentiation into multiple cell lineages. Moreover, ASCs secrete a wide range of growth factors that can stimulate tissue regeneration. Therefore, the clinical use of ASCs is feasible. However, the potential of ASCs differs depending on the donor's medical condition, including diseases such as diabetes. Recent studies demonstrated that ASCs from diabetic donors exhibit reduced proliferative potential and a smaller proportion of stem cell marker-positive cells. Therefore, to ensure the success of regenerative medicine, tissue engineering methods must be improved by the incorporation of factors that increase the proliferation and differentiation of stem/progenitor cells when autologous cells are used. Platelet-rich plasma (PRP), which contains high levels of diverse growth factors that can stimulate stem cell proliferation and cell differentiation in the context of tissue regeneration, has recently been identified as a biological material that could be applied to tissue regeneration. Thus, co-transplantation of ASCs and PRP represents a promising novel approach for cell therapy in regenerative medicine. In this review, we describe the potential benefits of adding PRP to ASCs and preclinical and clinical studies of this approach in various medical fields. We also discuss the mechanisms of PRP action and future cell-based therapies using co-transplantation of ASCs and PRP.
Collapse
Affiliation(s)
- Morikuni Tobita
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 1138421, Japan
| | - Satoshi Tajima
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 1138421, Japan
| | - Hiroshi Mizuno
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 1138421, Japan.
| |
Collapse
|
47
|
Lee MW, Ryu S, Kim DS, Sung KW, Koo HH, Yoo KH. Strategies to improve the immunosuppressive properties of human mesenchymal stem cells. Stem Cell Res Ther 2015; 6:179. [PMID: 26445096 PMCID: PMC4596374 DOI: 10.1186/s13287-015-0178-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of particular interest for the treatment of immune-related diseases because of their immunosuppressive capacities. However, few clinical trials of MSCs have yielded satisfactory results. A number of clinical trials using MSCs are currently in progress worldwide. Unfortunately, protocols and methods, including optimized culture conditions for the harvest of MSCs, have not been standardized. In this regard, complications in the ex vivo expansion of MSCs and MSC heterogeneity have been implicated in the failure of clinical trials. In this review, potential strategies to obtain MSCs with improved immunosuppressive properties and the potential roles of specific immunomodulatory genes, which are differentially upregulated in certain culture conditions, will be discussed.
Collapse
Affiliation(s)
- Myoung Woo Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Somi Ryu
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Dae Seong Kim
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Ki Woong Sung
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea
| | - Hong Hoe Koo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea. .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea.
| | - Keon Hee Yoo
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea. .,Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, 50 Irwon-Dong, Gangnam-Gu, Seoul, 135-710, Korea.
| |
Collapse
|
48
|
Wu Q, Ji FK, Wang JH, Nan H, Liu DL. Stromal cell-derived factor 1 promoted migration of adipose-derived stem cells to the wounded area in traumatic rats. Biochem Biophys Res Commun 2015; 467:140-5. [PMID: 26392311 DOI: 10.1016/j.bbrc.2015.09.097] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) were effective in treating wound. Stromal cell-derived factor-1 (SDF-1), a chemokine usually called CXCL12, is well known for its chemotaxis in induction of cell migration. However, little is known about the SDF-1responsible for the complex migration of ADSCs from residence to injured sites. OBJECTIVE Herein, we firstly showed SDF-1 is a major regulator involved in migration of ADSCs during wound repair in vivo. METHODS Trauma in rats was induced by surgical operation. The levels of SDF-1 in wounded tissue were assayed by ELISA. ADSCs were labeled with Green Fluorescent Protein (GFP), and then were transferred to injured rats by intracarotid injection. The plasma levels of ADSCs during wound healing were detected by flow cytometry, and ADSCs in injured tissue were evaluated by bioluminescence imaging in vivo and laser confocal microscopy (LCM), respectively. RESULTS ADSCs were successfully labeled with GFP. SDF-1 level reached to the peak value on 24 h after injury and then decreased continuously. Additionally, levels of plasma ADSCs in SDF-1 treated rats reached to the peak value (12%) at d21 after medicine delivery, while those of normal and injured rats showed the peak values of 6.28% and 9.84% at d7 and d21, respectively. Finally, the results of LCM indicated treatment of ectogenic SDF-1 obviously enhanced GFP-ADSCs distribution in wounded tissues. CONCLUSION Our results indicated that SDF-1 treatment obviously promoted the migration and directed distribution of ADSCs in traumatic tissue.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Fu-Kang Ji
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Jin-Huang Wang
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Hua Nan
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Da-Lie Liu
- Department of Plastic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
49
|
Zheng K, Wu W, Yang S, Huang L, Chen J, Gong C, Fu Z, Zhang L, Tan J. Bone marrow mesenchymal stem cell implantation for the treatment of radioactivity‑induced acute skin damage in rats. Mol Med Rep 2015; 12:7065-71. [PMID: 26323987 DOI: 10.3892/mmr.2015.4270] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 06/22/2015] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to observe the role of mesenchymal stem cells (MSCs) in the repair of acute skin damage caused by radiation. Rat bone marrow MSCs (BMSCs) were isolated and cultured in vitro. A rat model of radiation‑induced acute skin damage was established by irradiation of the hind legs of Sprague-Dawley rats using a linear accelerator (45 Gy). After irradiation, rats were randomly divided into two groups: BMSC group and control group. Rats in the BMSC group were treated with a tail vein injection of 2x106 BMSCs (1 ml) immediately after irradiation and a local multipoint injection of 2x106 BMSCs at the injured area two weeks later. Then the wound healing of each rat was observed. The expression of transforming growth factor (TGF)‑β1, stromal cell‑derived factor-1 (SDF‑1) and prostaglandin E2 (PGE2) in the wounded tissues was determined by immunohistochemistry. The results demonstrated that skin damage was milder in the BMSC group than in the control group. Moreover, the speed of healing in the BMSC group was better than that in the control group. In addition, the wound score, it was significantly lower in the BMSC group than in the control group (P<0.05). The expression of PGE2 and TGF‑β1 in the BMSC group was also significantly lower than that in the control group (P<0.05), whereas the SDF‑1 expression was significantly higher in the BMSC group than that in the control group (P<0.05). BMSCs can effectively reduce inflammation and fibrosis in the wounded skin and promote the repair of acute radioactive skin injury. Thus, may be developed as a novel treatment for wound healing.
Collapse
Affiliation(s)
- Kai Zheng
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Weizhen Wu
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Shunliang Yang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Lianghu Huang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Jin Chen
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Chungui Gong
- Radiotherapy Center, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Zhichao Fu
- Radiotherapy Center, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Linlin Zhang
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| | - Jianming Tan
- Cell and Organ Transplant Institute, Fuzhou General Hospital, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
50
|
Li Z, Wang Y, Xing H, Wang Z, Hu H, An R, Xu H, Liu Y, Liu B. Protective efficacy of intravenous transplantation of adipose-derived stem cells for the prevention of radiation-induced salivary gland damage. Arch Oral Biol 2015; 60:1488-96. [PMID: 26263537 DOI: 10.1016/j.archoralbio.2015.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 06/25/2015] [Accepted: 07/25/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE High-dose radiation therapy in the head and neck area can lead to irreversible damage to salivary glands (SGs) with consequent xerostomia. Adipose-derived stem cells (ADSCs) have been shown to repair or rescue damaged SGs. Thus, we investigated the protective efficacy of ADSCs in the prevention of SG damage induced by high dose radiation. METHODS Third-passage ADSCs (1×10(6)) were transplanted by intravenous infusion into the tail-vein of 8-week-old C57BL/6 mice, immediately after local irritation at a dose of 18Gy. The process was repeated twice a week during a period of six consecutive weeks. Eight weeks after radiation, functional evaluations were conducted by measuring salivary flow rate (SFR). Histological, immunohistochemical and transmission electron microscopic (TEM) examinations were performed to analyze microstructural and ultrastructural changes, microvessel density, amylase production, apoptosis, and proliferation activity. RESULTS Intravenously administrated ADSCs could home to irradiated SGs within 24h after infusion, significantly increasing SG weights, improving SFR, and preserving the microscopic morphologies of SGs eight weeks post-radiation. More functional acini, higher amylase production levels, and higher microvessel densities were observed in ADSC-treated SGs than in irradiated SGs. Additionally, enhanced cell proliferation activity and reduced radiation-induced SG apoptosis was observed in the ADSC-treated group when compared with the irradiated group. CONCLUSION Systemic administration of ADSCs immediately after radiation at a dose of 18Gy can protect both the morphology and function of SGs eight weeks after radiation in mice, and can be used as a protective measure for the prevention of SG damage induced by high-dose radiation.
Collapse
Affiliation(s)
- Zhijin Li
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yan Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Hongyan Xing
- Department of Stomatology, Xinzhou 2nd People's Hospital, 120 Yong Xing Nan Road, Xinzhou, Shanxi Province 034100, PR China
| | - Zhifa Wang
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Hanqing Hu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Ran An
- State Key Laboratory of Military Stomatology, Laboratory Animal Center, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Haiyan Xu
- State Key Laboratory of Military Stomatology, Laboratory Animal Center, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China
| | - Yanpu Liu
- State Key Laboratory of Military Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China.
| | - Bin Liu
- State Key Laboratory of Military Stomatology, Laboratory Animal Center, School of Stomatology, Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|