1
|
Guo X, Niu Z, Zhuang Y, Zhao Y, Ding Z, Shi J, Hou S, Fan H, Lv Q. Bone marrow mesenchymal stromal cells attenuate smoke inhalation injury by regulating the M1/M2-Th17/Treg immune homeostasis axis. Int Immunopharmacol 2024; 141:112986. [PMID: 39182266 DOI: 10.1016/j.intimp.2024.112986] [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: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
Smoke inhalation injury (SII) is the leading cause of death in fire burn patients. The inflammatory response induced by smoke inhalation is a significant factor in the development of acute lung injury or acute respiratory distress syndrome (ALI/ARDS). Mesenchymal stem cells (MSCs) can alleviate various inflammatory diseases by regulating the polarization of macrophages from the M1 to the M2 phenotype. Moreover, MSCs can facilitate the inflammatory response by regulating Th17/Treg homeostasis. However, little is known about the associations among MSCs, M1/M2 macrophages and Th17/Treg homeostasis. Therefore, the purpose of this study was to evaluate whether MSCs affect subsequent Th17/Treg differentiation and immune homeostasis by regulating M1/M2 polarization in SII. Our results showed that bone marrow mesenchymal stem cells (BMSCs) ameliorated lung inflammatory injury and fibrosis after SII by affecting the polarization of alveolar macrophages (AMs) from the M1 to the M2 phenotype. Moreover, BMSCs maintain Th17/Treg immune homeostasis by increasing the proportion of Treg cells and decreasing the proportion of Th17 cells. In vitro, we further demonstrated that BMSCs promoted the polarization of AMs from the M1 to the M2 phenotype and decreased IL-23 levels. Reduced IL-23 decreased Th17 differentiation and promoted Th17/Treg balance. Therefore, BMSCs ameliorate the inflammatory response and lung damage after SII through regulating M1/M2 polarization and subsequent Th17/Treg immune homeostasis, which are linked to alveolar macrophage-derived IL-23. These findings provide novel insight into how BMSCs regulate the M1/M2-Th17/Treg immune homeostasis axis and provide new therapeutic targets for more effective control of the inflammatory response after SII.
Collapse
Affiliation(s)
- Xiaoqin Guo
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Zhifang Niu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Yong Zhuang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Yunlong Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Ziling Ding
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Jie Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China.
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China.
| | - Qi Lv
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, China; Wenzhou Safety (Emergency) Institute, Tianjin University, Wenzhou 325026, China.
| |
Collapse
|
2
|
Kang D, Kim T, Choi GE, Park A, Yoon J, Yu J, Suh N. miR-29a-3p orchestrates key signaling pathways for enhanced migration of human mesenchymal stem cells. Cell Commun Signal 2024; 22:365. [PMID: 39020373 PMCID: PMC11256664 DOI: 10.1186/s12964-024-01737-0] [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: 04/16/2024] [Accepted: 07/04/2024] [Indexed: 07/19/2024] Open
Abstract
BACKGROUND The homing of human mesenchymal stem cells (hMSCs) is crucial for their therapeutic efficacy and is characterized by the orchestrated regulation of multiple signaling modules. However, the principal upstream regulators that synchronize these signaling pathways and their mechanisms during cellular migration remain largely unexplored. METHODS miR-29a-3p was exogenously expressed in either wild-type or DiGeorge syndrome critical region 8 (DGCR8) knockdown hMSCs. Multiple pathway components were analyzed using Western blotting, immunohistochemistry, and real-time quantitative PCR. hMSC migration was assessed both in vitro and in vivo through wound healing, Transwell, contraction, and in vivo migration assays. Extensive bioinformatic analyses using gene set enrichment analysis and Ingenuity pathway analysis identified enriched pathways, upstream regulators, and downstream targets. RESULTS The global depletion of microRNAs (miRNAs) due to DGCR8 gene silencing, a critical component of miRNA biogenesis, significantly impaired hMSC migration. The bioinformatics analysis identified miR-29a-3p as a pivotal upstream regulator. Its overexpression in DGCR8-knockdown hMSCs markedly improved their migration capabilities. Our data demonstrate that miR-29a-3p enhances cell migration by directly inhibiting two key phosphatases: protein tyrosine phosphatase receptor type kappa (PTPRK) and phosphatase and tensin homolog (PTEN). The ectopic expression of miR-29a-3p stabilized the polarization of the Golgi apparatus and actin cytoskeleton during wound healing. It also altered actomyosin contractility and cellular traction forces by changing the distribution and phosphorylation of myosin light chain 2. Additionally, it regulated focal adhesions by modulating the levels of PTPRK and paxillin. In immunocompromised mice, the migration of hMSCs overexpressing miR-29a-3p toward a chemoattractant significantly increased. CONCLUSIONS Our findings identify miR-29a-3p as a key upstream regulator that governs hMSC migration. Specifically, it was found to modulate principal signaling pathways, including polarization, actin cytoskeleton, contractility, and adhesion, both in vitro and in vivo, thereby reinforcing migration regulatory circuits.
Collapse
Affiliation(s)
- Dayeon Kang
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Taehwan Kim
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Ga-Eun Choi
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea
| | - Arum Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jin Yoon
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Jinho Yu
- Department of Pediatrics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Nayoung Suh
- Department of Medical Sciences, General Graduate School, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
- Department of Pharmaceutical Engineering, College of Medical Sciences, Soon Chun Hyang University, Asan, 31538, Republic of Korea.
| |
Collapse
|
3
|
Jiang Z, Yu J, Zhou H, Feng J, Xu Z, Wan M, Zhang W, He Y, Jia C, Shao S, Guo H, Liu B. Research hotspots and emerging trends of mesenchymal stem cells in cardiovascular diseases: a bibliometric-based visual analysis. Front Cardiovasc Med 2024; 11:1394453. [PMID: 38873270 PMCID: PMC11169657 DOI: 10.3389/fcvm.2024.1394453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) have important research value and broad application prospects in cardiovascular diseases (CVDs). However, few bibliometric analyses on MSCs in cardiovascular diseases are available. This study aims to provide a thorough review of the cooperation and influence of countries, institutions, authors, and journals in the field of MSCs in cardiovascular diseases, with the provision of discoveries in the latest progress, evolution paths, frontier research hotspots, and future research trends in the regarding field. Methods The articles related to MSCs in cardiovascular diseases were retrieved from the Web of Science. The bibliometric study was performed by CiteSpace and VOSviewer, and the knowledge map was generated based on data obtained from retrieved articles. Results In our study, a total of 4,852 publications launched before August 31, 2023 were accessed through the Web of Science Core Collection (WoSCC) database via our searching strategy. Significant fluctuations in global publications were observed in the field of MSCs in CVDs. China emerged as the nation with the largest number of publications, yet a shortage of high-quality articles was noted. The interplay among countries, institutions, journals and authors is visually represented in the enclosed figures. Importantly, current research trends and hotspots are elucidated. Cluster analysis on references has highlighted the considerable interest in exosomes, extracellular vesicles, and microvesicles. Besides, keywords analysis revealed a strong emphasis on myocardial infarction, therapy, and transplantation. Treatment methods-related keywords were prominent, while keywords associated with extracellular vesicles gathered significant attention from the long-term perspective. Conclusion MSCs in CVDs have become a topic of active research interest, showcasing its latent value and potential. By summarizing the latest progress, identifying the research hotspots, and discussing the future trends in the advancement of MSCs in CVDs, we aim to offer valuable insights for considering research prospects.
Collapse
Affiliation(s)
- Zhihang Jiang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Yu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houle Zhou
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zehui Xu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Melisandre Wan
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing He
- Department of Preventive Medicine, College of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyao Jia
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Shuijin Shao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haidong Guo
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Dantas JR, Araujo DB, Silva KR, Souto DL, Pereira MDFC, Raggio LR, Claudio-da Silva C, Couri CE, Maiolino A, Rebellato CLK, Daga DR, Senegaglia AC, Brofman PRS, Baptista LS, Oliveira JEPD, Zajdenverg L, Rodacki M. Adipose Tissue-Derived Stromal/Stem Cells Transplantation with Cholecalciferol Supplementation in Recent-Onset Type 1 Diabetes Patients: Twelve Months Follow-Up. Horm Metab Res 2023; 55:536-545. [PMID: 37192655 DOI: 10.1055/a-2094-1039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
To evaluate safety and therapeutic effect along 12 months of allogenic adipose tissue-derived stromal/stem cells (ASCs) transplantation with cholecalciferol (VITD) in patients with recent-onset type 1 diabetes (T1D). Prospective, phase II, open trial, pilot study in which patients with recent onset T1D received ASCs (1xKgx106 cells) and VITD 2000UI/day for 12 months (group 1) and were compared to controls with standard insulin therapy (group 2). Adverse events, C-peptide area under the curve (CPAUC), insulin dose, HbA1c and frequency of FoxP3+ in CD4+ or CD8+ T-cells(flow cytometry) were evaluated at baseline(T0), after 3(T3), 6(T6) and 12 months(T12). Eleven patients completed follow up (7:group 1;4:group 2). Group 1 had lower insulin requirement at T3(0.24±0.18vs0.53±0.23UI/kg,p=0.04), T6(0.24±0.15vs0.66±0.33 UI/kg,p=0.04) and T12(0.39±0.15vs0.74±0.29 UI/Kg,p=0.04).HbA1c was lower at T6 (50.57±8.56vs72.25±10.34 mmol/mol,p=0.01), without differences at T12 (57.14±11.98 in group 1 vs. 73.5±14.57 mmol/min in group 2, p=0.16). CPAUC was not significantly different between groups at T0(p=0.07), higher in group 1 at T3(p=0.04) and T6(p=0.006), but similar at T12(p=0.23). IDAA1c was significantly lower in group 1 than group 2 at T3,T6 and T12 (p=0.006, 0.006 and 0.042, respectively). IDDA1c was inversely correlated to FoxP3 expression in CD4 and CD8+ T cells at T6 (p<0.001 and p=0.01, respectively). In group 1, one patient had recurrence of a benign teratoma that was surgically removed, not associated to the intervention. ASCs with VITD without immunosuppression were safe and associated lower insulin requirements, better glycemic control, and transient better pancreatic function in recent onset T1D, but the potential benefits were not sustained.
Collapse
Affiliation(s)
- Joana R Dantas
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora Batista Araujo
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Ribeiro Silva
- Laboratory of Tissue Bioengineering, Instituto Nacional de Metrologia Qualidade e Tecnologia Campus de Xerem, Duque de Caxias, Brazil
- Histology and Embryology Departament, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Debora Lopes Souto
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Luiz Ronir Raggio
- Institute of Public Health Studies, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos Eduardo Couri
- Internal Medicine, Universidade de São Paulo Faculdade de Medicina de Ribeirão Preto, Ribeirao Preto, Brazil
| | - Angelo Maiolino
- Hematology Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Debora Regina Daga
- Core Cell Technology, Pontifical Catholic University of Parana, Curitiba, Brazil
| | | | | | - Leandra S Baptista
- Laboratory of Tissue Bioengineering, Instituto Nacional de Metrologia Qualidade e Tecnologia Campus de Xerem, Duque de Caxias, Brazil
- Center for Biological Research (Numpex-Bio), Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Lenita Zajdenverg
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Melanie Rodacki
- Nutrology and Diabetes Department, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Wang J, Tian J, Wang L, Yang ZW, Xu P. Mesenchymal stem cells regulate M1 polarization of peritoneal macrophages through the CARD9-NF-κB signaling pathway in severe acute pancreatitis. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2023; 30:338-350. [PMID: 35738898 DOI: 10.1002/jhbp.1205] [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: 02/09/2022] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Macrophages release large numbers of proinflammatory cytokines that trigger inflammatory cascade reactions, which promote the rapid development of severe acute pancreatitis (SAP) from local to systemic inflammation. The ability of mesenchymal stem cells (MSCs) to suppress inflammation is related to inhibition of M1 polarization of macrophages. Our previous studies revealed that caspase recruitment domain protein 9 (CARD9) was involved in SAP inflammation and activation of the CARD9-NF-κB signaling pathway plays an important proinflammatory role in SAP. At present, there is no effective treatment to control the inflammatory response in SAP. Therefore, the aim of the present study was to determine whether MSCs regulate the polarization of macrophages through the CARD9-NF-κB signaling pathway in SAP. METHODS Short hairpin RNA interference technology and coculture in vitro were used to assess the activation status of the CARD9-NF-κB signal pathway in macrophages. Furthermore, flow cytometry was used to determine the polarization state of macrophages. RESULTS The results showed MSCs inhibited CARD9 expression in vivo and in vitro (P < .05), alleviated inflammation induced by proinflammatory cytokines, and inhibited the phosphorylation of NF-κB in macrophages both in vivo and in vitro. Meanwhile, MSCs downregulated the CARD9-NF-κB signal pathway and inhibited M1 polarization of macrophages. CONCLUSION In conclusion, MSCs regulate M1 polarization of peritoneal macrophages through the CARD9-NF-κB signaling pathway in SAP and transplantation of MSCs presents an effective treatment option for SAP.
Collapse
Affiliation(s)
- Jing Wang
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
- Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Jun Tian
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
- Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Lin Wang
- Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhi-Wen Yang
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
- Shanghai Songjiang Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ping Xu
- Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Preparatory Stage), Shanghai, China
| |
Collapse
|
6
|
Yin Q, Zou T, Sun S, Yang D. Cell therapy for neuropathic pain. Front Mol Neurosci 2023; 16:1119223. [PMID: 36923653 PMCID: PMC10008860 DOI: 10.3389/fnmol.2023.1119223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/07/2023] [Indexed: 03/02/2023] Open
Abstract
Neuropathic pain (NP) is caused by a lesion or a condition that affects the somatosensory system. Pathophysiologically, NP can be ascribed to peripheral and central sensitization, implicating a wide range of molecular pathways. Current pharmacological and non-pharmacological approaches are not very efficacious, with over half of NP patients failing to attain adequate pain relief. So far, pharmacological and surgical treatments have focused primarily on symptomatic relief by modulating pain transduction and transmission, without treating the underlying pathophysiology. Currently, researchers are trying to use cell therapy as a therapeutic alternative for the treatment of NP. In fact, mounting pre-clinical and clinical studies showed that the cell transplantation-based therapy for NP yielded some encouraging results. In this review, we summarized the use of cell grafts for the treatment of NP caused by nerve injury, synthesized the latest advances and adverse effects, discussed the possible mechanisms to inform pain physicians and neurologists who are endeavoring to develop cell transplant-based therapies for NP and put them into clinical practice.
Collapse
Affiliation(s)
- QingHua Yin
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - TianHao Zou
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - ShuJun Sun
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dong Yang
- Department of Pain, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Shi L, Zhang Z, Deng M, Zheng F, Liu W, Ye S. Biological mechanisms and applied prospects of mesenchymal stem cells in premature ovarian failure. Medicine (Baltimore) 2022; 101:e30013. [PMID: 35960112 PMCID: PMC9371578 DOI: 10.1097/md.0000000000030013] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/05/2022] [Accepted: 06/06/2022] [Indexed: 01/04/2023] Open
Abstract
Premature ovarian failure (POF), also known as primary ovarian insufficiency (POI), refers to the loss of ovarian function in women after puberty and before the age of 40 characterized by high serum gonadotropins and low estrogen, irregular menstruation, amenorrhea, and decreased fertility. However, the specific pathogenesis of POF is unexplained, and there is no effective therapy for its damaged ovarian tissue structure and reduced reserve function. Mesenchymal stem cells (MSCs), with multidirectional differentiation potential and self-renewal ability, as well as the cytokines and exosomes they secrete, have been studied and tested to play an active therapeutic role in a variety of degenerative pathologies, and MSCs are the most widely used stem cells in regenerative medicine. MSCs can reverse POI and enhance ovarian reserve function through differentiation into granulosa cells (GCs), immune regulation, secretion of cytokines and other nutritional factors, reduction of GCs apoptosis, and promotion of GCs regeneration. Many studies have proved that MSCs may have a restorative effect on the structure and fertility of injured ovarian tissues and turn to be a useful clinical approach to the treatment of patients with POF in recent years. We intend to use MSCs-based therapy to completely reverse POI in the future.
Collapse
Affiliation(s)
- Lan Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Zhifen Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Miao Deng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Fangyuan Zheng
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Wenhua Liu
- Department of Obstetrics and Gynecology, Hangzhou Women’s Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang Province, People’s Republic of China
| | - Shujin Ye
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
8
|
Nir D, Ribarski-Chorev I, Shimoni C, Strauss C, Frank J, Schlesinger S. Antioxidants Attenuate Heat Shock Induced Premature Senescence of Bovine Mesenchymal Stem Cells. Int J Mol Sci 2022; 23:ijms23105750. [PMID: 35628565 PMCID: PMC9147428 DOI: 10.3390/ijms23105750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/15/2022] [Accepted: 05/18/2022] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSC) have many roles that are important for the body's proper functioning. When the MSC pool is damaged, it is often correlated with impaired development or health of the organism. MSC are known for their anti-inflammatory, immunomodulatory and trophic characteristics that play an important role in the physiological homeostasis of many tissues. Heat shock impairs MSC capacity by inducing the generation of reactive oxygen species and mitochondrial dysfunction, which, in turn, send the cells into a state of premature senescence. Here, we pre-exposed MSC to melatonin, resveratrol, or curcumin, which are natural antioxidative compounds, and tested the protective effects of these substances from oxidative stress and aging. Our data showed that pre-exposure of MSC to antioxidants decreased reactive oxygen species while mitochondrial damage remained high. Additionally, although the proliferation of the cells was slow, antioxidants protected the cells from premature senescence, and subsequent cytokine release was prevented. We conclude that while elevated temperatures directly cause mitochondrial damage, senescence is induced by elevated ROS levels. We suggest that heat shock alters cell and tissue homeostasis by several independent mechanisms; however, reducing tissue senescence will reduce damage and provide a pathway to overcome physiological challenges in animals.
Collapse
Affiliation(s)
- Dana Nir
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (D.N.); (I.R.-C.); (C.S.); (C.S.)
| | - Ivana Ribarski-Chorev
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (D.N.); (I.R.-C.); (C.S.); (C.S.)
| | - Chen Shimoni
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (D.N.); (I.R.-C.); (C.S.); (C.S.)
| | - Carmit Strauss
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (D.N.); (I.R.-C.); (C.S.); (C.S.)
| | - Jan Frank
- Department of Food Biofunctionality, Institute of Nutritional Sciences, University of Hohenheim, D-70599 Stuttgart, Germany;
| | - Sharon Schlesinger
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food, and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (D.N.); (I.R.-C.); (C.S.); (C.S.)
- Correspondence:
| |
Collapse
|
9
|
Kim EY, Kim HS, Hong KS, Chung HM, Park SP, Noh G. Mesenchymal stem/stromal cell therapy in atopic dermatitis and chronic urticaria: immunological and clinical viewpoints. Stem Cell Res Ther 2021; 12:539. [PMID: 34635172 PMCID: PMC8503727 DOI: 10.1186/s13287-021-02583-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/30/2021] [Indexed: 12/29/2022] Open
Abstract
Allergic diseases are immune-mediated diseases. Allergies share a common immunopathogenesis, with specific differences according to the specific disease. Mesenchymal stem/stromal cells (MSCs) have been applied to people suffering from allergic and many other diseases. In this review, the immunologic roles of MSCs are systemically reviewed according to disease immunopathogenesis from a clinical viewpoint. MSCs seem to be a promising therapeutic modality not only as symptomatic treatments but also as causative and even preventive treatments for allergic diseases, including atopic dermatitis and chronic urticaria.
Collapse
Affiliation(s)
| | - Hyuk Soon Kim
- Department of Biomedical Sciences, College of Natural Science, The Graduate School of Dong-A University, Busan, Korea.,Department of Health Sciences, The Graduate School of Dong-A University, Busan, Korea
| | | | - Hyung-Min Chung
- Miraecellbio Co., Ltd., Seoul, Korea.,Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul, Korea
| | - Se-Pill Park
- Miraecellbio Co., Ltd., Seoul, Korea. .,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, 63243, Korea.
| | - Geunwoong Noh
- Department of Allergy, Allergy and Clinical Immunology Center, Cheju Halla General Hospital, Doreongno 65, Jeju-si, 63127, Jeju Special Self-Governing Province, Korea.
| |
Collapse
|
10
|
Abu Almaaty AH, Elmasry RA, Farrag MS, Althobaiti F, Aldhahrani A, Fayad E, Hussain MA. Impact of Human Umbilical Cord Blood Mononuclear Cells on Gentamicin-Induced Renal Injury and Genotoxicity in Rats. Front Med (Lausanne) 2021; 8:689691. [PMID: 34490290 PMCID: PMC8417307 DOI: 10.3389/fmed.2021.689691] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/05/2021] [Indexed: 11/21/2022] Open
Abstract
Background: Acute kidney injury (AKI), also known as acute renal failure (ARF), has received considerable critical attention in recent years. Gentamicin (GM) is an antibiotic whose prolonged use results in AKI with a high mortality rate. Methods: Fifty adult female albino rats weighing 150–200 g were used. The animals were divided into five groups: the first group was the normal healthy control one, the second group received only 1 × 106 HUCB mononuclear cells (MNCs)/rat by intravenous (iv) injection, the third diseased group was given GM 100 mg/kg for 10 consecutive days by intraperitoneal injections, the fourth preventive group received 1 × 106 HUCB MNCs/rat by iv injection 24 h before gentamicin treatment, and the fifth treated group received 1 × 106 HUCB MNCs/rat by iv injection 24 h after gentamicin treatment. After 1 week of treatment, blood samples were collected, and kidneys were removed for histopathological examination. Results: Rats treated with HUCB MNCs in the treated group had a significant decrease in renal damage, low levels of biomarkers for nephrotoxicities such as serum creatinine and blood urea nitrogen, and low chromosomal aberrations compared to the diseased third group. The gene expression of KIM-1 and NGAL was decreased in response to HUCB treatment. Conclusions: HUCB MNCs have a curative effect against AKI and gentamicin-induced genotoxicity owing to their regenerative property.
Collapse
Affiliation(s)
- Ali H Abu Almaaty
- Department of Zoology, Faculty of Science, Port Said University, Port Said, Egypt
| | - Reham A Elmasry
- Department of Zoology, Faculty of Science, Port Said University, Port Said, Egypt
| | - Mayada S Farrag
- Department of Pathology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Fayez Althobaiti
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif, Saudi Arabia
| | - Adil Aldhahrani
- Clinical Laboratory Sciences Department, Turabah University Faculty, Taif University, Taif, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, Faculty of Sciences, Taif University, Taif, Saudi Arabia
| | - Mona A Hussain
- Department of Physiology, Faculty of Medicine, Port Said University, Port Said, Egypt
| |
Collapse
|
11
|
Ho CT, Wu MH, Chen MJ, Lin SP, Yen YT, Hung SC. Combination of Mesenchymal Stem Cell-Delivered Oncolytic Virus with Prodrug Activation Increases Efficacy and Safety of Colorectal Cancer Therapy. Biomedicines 2021; 9:548. [PMID: 34068264 PMCID: PMC8153168 DOI: 10.3390/biomedicines9050548] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Although oncolytic viruses are currently being evaluated for cancer treatment in clinical trials, systemic administration is hindered by many factors that prevent them from reaching the tumor cells. When administered systemically, mesenchymal stem cells (MSCs) target tumors, and therefore constitute good cell carriers for oncolytic viruses. MSCs were primed with trichostatin A under hypoxia, which upregulated the expression of CXCR4, a chemokine receptor involved in tumor tropism, and coxsackievirus and adenovirus receptor that plays an important role in adenoviral infection. After priming, MSCs were loaded with conditionally replicative adenovirus that exhibits limited proliferation in cells with a functional p53 pathway and encodes Escherichia coli nitroreductase (NTR) enzymes (CRAdNTR) for targeting tumor cells. Primed MSCs increased tumor tropism and susceptibility to adenoviral infection, and successfully protected CRAdNTR from neutralization by anti-adenovirus antibodies both in vitro and in vivo, and specifically targeted p53-deficient colorectal tumors when infused intravenously. Analyses of deproteinized tissues by UPLC-MS/QTOF revealed that these MSCs converted the co-administered prodrug CB1954 into cytotoxic metabolites, such as 4-hydroxylamine and 2-amine, inducing oncolysis and tumor growth inhibition without being toxic for the host vital organs. This study shows that the combination of oncolytic viruses delivered by MSCs with the activation of prodrugs is a new cancer treatment strategy that provides a new approach for the development of oncolytic viral therapy for various cancers.
Collapse
Affiliation(s)
- Chun-Te Ho
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| | - Mei-Hsuan Wu
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (M.-H.W.); (S.-P.L.)
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital & Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan;
| | - Shih-Pei Lin
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; (M.-H.W.); (S.-P.L.)
| | - Yu-Ting Yen
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| | - Shih-Chieh Hung
- Drug Development Center, Institute of New Drug Development, Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung 404, Taiwan; (C.-T.H.); (Y.-T.Y.)
- Integrative Stem Cell Center, Department of Orthopaedics, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
12
|
Yang L, Dai H, Yang J, Yang H, Yang D, Kang J. Molecular Mechanism of Neurotrophic Factor-Activated Long Non-Coding RNA Plasmacytoma Variant Translocation 1 Promoting Mesenchymal Stem Cell Migration and Repair of Fractures. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
It has been reported that neurotrophic factor (NF) promotes bone marrow mesenchymal stem cells (MSCs) migration to repair fractures. However, whether and how lncRNA PVT1 regulates differentiation induced by neurotrophic factors to promote MSC migration to repair fractures has not been
explored. To explore the molecular mechanism of neurotrophic factor activating lncRNA PVT1 to promote MSC migration and repair fractures. Differential expression of neurotrophic factors stimulated by MSCs was analyzed based on microarray lncRNA and lncRNAs was further verified by qRT-PCR.
The conditions of promoting MSC migration and osteogenic differentiation were identified by trans-fection of lncRNA PVT1 overexpressed plasmids and inhibitor and the targets of its regulation were confirmed by target gene prediction tools. In this study lncRNA array and qRT-PCR showed that
lncRNA PVT1 was significantly down-regulated during neurotrophic factor-induced MSCs differentiation. Transfection of lncRNA PVT1 overexpression plasmid significantly inhibited the expression of osteogenic markers alkaline phosphatase (ALP) and osteopontin (OPN) in MSCs, while transfection
of lncRNA PVT1 inhibitor promoted the expression of alkaline phosphatase (ALP) and osteopontin (OPN). lncRNA PVT1 is a negative regulator of MSCs differentiation induced by neurotrophic factors. The distal deletion homologous box 5(DLX5) was identified as the target of lncRNA PVT1 and the
relationship between lncRNA PVT1 inhibiting the expression of DLX5 and the osteogenic differentiation of MSCs was verified in MSCs. lncRNA PVT1 negatively regulates the migration and differentiation of MSCs induced by neurotrophic factors by targeting DLX5, providing the foundation for bone
repair.
Collapse
Affiliation(s)
- Linyu Yang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Haoping Dai
- Hospital (t.c.m) Affiliated to Southwest Medical University-Spine Surgery, Luzhou, Sichuan, 646000, China
| | - Jian Yang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Han Yang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Daoyin Yang
- Luzhou Jiuzheng Orthopaedic Hospital, Luzhou, Sichuan, 646099, China
| | - Jianping Kang
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
13
|
Comparative study of commercial protocols for high recovery of high-purity mesenchymal stem cell-derived extracellular vesicle isolation and their efficient labeling with fluorescent dyes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 35:102396. [PMID: 33864911 DOI: 10.1016/j.nano.2021.102396] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 01/01/2023]
Abstract
The extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) can be used as carriers for therapeutic molecules and drugs to target disordered tissues. This aimed to compare the protocols used for isolation of MSC-derived EVs by comparing EV collection conditions and three commercial purification kits. We also determined appropriate fluorescent dyes for labeling EVs. MSC-derived EVs were efficiently secreted during cell growth and highly purified by the phosphatidyl serine-based affinity kit. Although the EV membrane was more efficiently labeled with the fluorescent dye PKH67 compared to other probes, the efficiency was not enough to accurately analyze the endothelial cellular uptake of EVs. Results verified the easy protocol for isolating and fluorescently labeling EVs with commercial reagents and kits, but meanwhile, further modification of the protocol is required in order to scale up the amount of EVs derived from MSCs using fluorescent probes.
Collapse
|
14
|
Su Y, Zhang T, Huang T, Gao J. Current advances and challenges of mesenchymal stem cells-based drug delivery system and their improvements. Int J Pharm 2021; 600:120477. [PMID: 33737099 DOI: 10.1016/j.ijpharm.2021.120477] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have recently emerged as a promising living carrier for targeted drug delivery. A wealth of literature has shown evidence for great advances in MSCs-based drug delivery system (MSCs-DDS) in the treatment of various diseases. Nevertheless, as this field of study rapidly advances, several challenges associated with this delivery strategy have arisen, mainly due to the inherent limitations of MSCs. To this end, several novel technologies are being developed in parallel to improve the efficiency or safety of this system. In this review, we introduce recent advances and summarize the present challenges of MSCs-DDS. We also highlight some potential technologies to improve MSCs-DDS, including nanotechnology, genome engineering technology, and biomimetic technology. Finally, prospects for application of artificially improved MSCs-DDS are addressed. The technologies summarized in this review provide a general guideline for the improvement of MSCs-DDS.
Collapse
Affiliation(s)
- Yuanqin Su
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ting Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China; Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
15
|
Wei X, Zheng Y, Zhang W, Tan J, Zheng H. Ultrasound‑targeted microbubble destruction‑mediated Galectin‑7‑siRNA promotes the homing of bone marrow mesenchymal stem cells to alleviate acute myocardial infarction in rats. Int J Mol Med 2020; 47:677-687. [PMID: 33416139 PMCID: PMC7797467 DOI: 10.3892/ijmm.2020.4830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are accepted as a form of cellular therapy to improve cardiac function following acute myocardial infarction (AMI). The present study was performed to investigate the synergistic effect of ultrasound-targeted microbubble destruction (UTMD)-mediated Galectin-7-small interfering (si)RNA with the homing of BMSCs for AMI. The rat model of AMI was established, followed by identification of BMSCs. Rats with AMI received BMSC transplantation, BMSC transplantation + UTMD + siRNA negative control, or BMSC transplantation + UTMD + Galectin-7-siRNA. The cardiac function, hemodynamics indexes, degree of myocardial fiber injury and expression of apoptosis-related proteins in myocardial tissues of rats were detected. The homing of BMSCs was observed, and the indexes of myocardial microenvironment and the TGF-β/Smads pathway-related proteins in myocardial tissues were determined. AMI rats treated with UTMD-mediated Galectin-7-siRNA exhibited improved cardiac function and hemodynamics-related indices, decreased myocardial fiber injury and apoptotic cells, as well as enhanced homing ability of BMSCs, improved myocardial microenvironment, and suppressed TGF-β1/Smads pathway activation. In conclusion, the present study demonstrated that UTMD-mediated Galectin-7-siRNA treatment could enhance the homing ability of BMSCs, thus alleviating AMI in rats.
Collapse
Affiliation(s)
- Xin Wei
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Yan Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Weilin Zhang
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Jing Tan
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Hong Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| |
Collapse
|
16
|
Chen J, Jiang J, Wang W, Qin J, Chen J, Chen W, Wang Y. Low intensity pulsed ultrasound promotes the migration of bone marrow- derived mesenchymal stem cells via activating FAK-ERK1/2 signalling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 47:3603-3613. [PMID: 31468983 DOI: 10.1080/21691401.2019.1657878] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To investigate the promoting effects and mechanisms of low intensity pulsed ultrasound (LIPUS) on the migration of bone marrow-derived mesenchymal stem cells (BMSCs). The BMSCs migration was researched from cell and animal experiments. In the cell experiment, the BMSCs was treated using LIPUS (30 mW/cm2, 20 min/day, 2 days), and the wound healing and transwell migration were observed. In the animal experiment, the BMSCs labelled with green fluorescent protein (GFP) were injected into rats with femoral defects via the tail vein (1 × 106/mL). The healing of bone was detected using x-ray and sampled for hematoxylin & eosin (H&E) staining and fluorescence microscopy. About the mechanisms, the cellular F-actin of cytoskeleton was stained with FITC-phalloidin. The changes of BMSCs genes after LIPUS treatment were screened using microarray assay and verified using quantitative real-time polymerase chain reaction (qRT-PCR). The biological processes of those genes were predicted by KEGG analysis. The protein expression levels of FAK, ERK1/2 and myosin II related migration were detected using western blotting. The results showed LIPUS promoted the BMSCs migration (p < .05) without significant temperature changes (p > .05) in vitro and in vivo than control group (p < .05). The cytoskeletal rearrangement was carried out, and the ITGA8 gene related with cell migration was found with high expression after LIPUS treatment (p < .05). FAK inhibitor (PF-573228) and ERK1/2 inhibitor (U0126) were proved, in turn, decreased the BMSCs migration induced using LIPUS (p < .05). LIPUS can promote the BMSCs migration in vitro and in vivo, one mechanism may be related to the activation of FAK-ERK1/2 signalling pathways using LIPUS.
Collapse
Affiliation(s)
- Junlin Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| | - Jingwei Jiang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| | - Wei Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| | - Juan Qin
- Guizhou Maternal and Child Health Hospital, Guizhou Medical University , Guizhou , China
| | - Jinyun Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| | - Wenzhi Chen
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| | - Yan Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing, the Ministry of Science and Technology, College of Biomedical Engineering, Chongqing Key Laboratory of Biomedical Engineering, Chongqing Collaborative Innovation Center for Minimally-Invasive and Noninvasive Medicine, Chongqing Medical University , Chongqing , China
| |
Collapse
|
17
|
Kamal MM, Kassem DH. Therapeutic Potential of Wharton's Jelly Mesenchymal Stem Cells for Diabetes: Achievements and Challenges. Front Cell Dev Biol 2020; 8:16. [PMID: 32064260 PMCID: PMC7000356 DOI: 10.3389/fcell.2020.00016] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is an alarming metabolic disease in which insulin secreting β-cells are damaged to various extent. Unfortunately, although currently available treatments help to manage the disease, however, patients usually develop complications, as well as decreased life quality and increased mortality. Thus, efficient therapeutic interventions to treat diabetes are urgently warranted. During the past years, mesenchymal stem cells (MSCs) have made their mark as a potential weapon in various regenerative medicine applications. The main fascination about MSCs lies in their potential to exert reparative effects on an amazingly wide spectrum of tissue injury. This is further reinforced by their ease of isolation and large ex vivo expansion capacity, as well as demonstrated multipotency and immunomodulatory activities. Among all the sources of MSCs, those isolated from umbilical cord-Wharton's jelly (WJ-MSCs), have been proved to provide a great source of MSCs. WJ-MSCs do not impose any ethical concerns as those which exist regarding ESCs, and represent a readily available non-invasive source, and hence suggested to become the new gold standard for MSC-based therapies. In the current review, we shall overview achievements, as well as challenges/hurdles which are standing in the way to utilize WJ-MSCs as a novel efficient therapeutic modality for DM.
Collapse
Affiliation(s)
- Mohamed M. Kamal
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- The Center for Drug Research and Development, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dina H. Kassem
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
18
|
Munir F, Jamshed MB, Shahid N, Muhammad SA, Ghanem NB, Qiyu Z. Current status of diagnosis and Mesenchymal stem cells therapy for acute pancreatitis. Physiol Rep 2019; 7:e14170. [PMID: 31691545 PMCID: PMC6832003 DOI: 10.14814/phy2.14170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/28/2019] [Accepted: 06/05/2019] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is an acute gastrointestinal disorder that is the most common and requiring emergency hospitalization. Its incidence is increasing worldwide, thus increasing the burden of medical services. Approximately 20% of the patients develop moderate to severe necrotizing pancreatitis associated with pancreatic or peri-pancreatic tissue necrosis and multiple organ failure. There are many reports about the anti-inflammatory effect of mesenchymal stem cells (MSCs) on pancreatitis and the repair of tissue damage. MSCs cells come from a wide range of sources, autologous MSCs come from bone marrow and allogeneic MSCs such as umbilical cord blood MSCs, placenta-derived MSCs, etc. The wide source is not only an advantage of MSCs but also a disadvantage of MSCs. Because of different cell sources and different methods of collection and preparation, it is impossible to establish a unified standard method for evaluation of efficacy. The biggest advantage of iMSCs is that it can be prepared by a standardized process, and can be prepared on a large scale, which makes it easier to commercialize. This paper reviews the present status of diagnosis and progress of MSCs therapy for AP.
Collapse
Affiliation(s)
- Fahad Munir
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouPeople’s Republic of China
| | - Muhammad B. Jamshed
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouPeople’s Republic of China
| | - Numan Shahid
- Department of General SurgeryThe School of International Studies of Wenzhou Medical UniversityWenzhouPeople’s Republic of China
| | - Syed A. Muhammad
- Institute of Molecular Biology and BiotechnologyBahaudin Zakariya UniversityMultan, PunjabPakistan
| | - Noor B. Ghanem
- The School of International Studies of Wenzhou Medical UniversityWenzhouPeople’s Republic of China
| | - Zhang Qiyu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouPeople’s Republic of China
| |
Collapse
|
19
|
Spiegel JL, Hambrecht M, Kohlbauer V, Haubner F, Ihler F, Canis M, Schilling AF, Böker KO, Dressel R, Streckfuss-Bömeke K, Jakob M. Radiation-induced sensitivity of tissue-resident mesenchymal stem cells in the head and neck region. Head Neck 2019; 41:2892-2903. [PMID: 31017352 DOI: 10.1002/hed.25768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/03/2019] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tissue-resident mesenchymal stem cells (MSCs) possess the ability to migrate to areas of inflammation and promote the regeneration of damaged tissue. However, it remains unclear how radiation influences this capacity of MSC in the head and neck region. METHODS Two types of MSCs of the head and neck region (mucosa [mMSC] and parotid gland [pMSC]) were isolated, cultured and exposed to single radiation dosages of 2 Gy/day up to 10 days. Effects on morphology, colony forming ability, apoptosis, chemokine receptor expression, cytokine secretion, and cell migration were analyzed. RESULTS Although MSC preserved MSC-specific regenerative abilities and immunomodulatory properties following irradiation in our in vitro model, we found a deleterious impact on colony forming ability, especially in pMSC. CONCLUSIONS MSC exhibited robustness and activation upon radiation for the support of tissue regeneration, but lost their potential to replicate, thus possibly leading to depletion of the local MSC-pool after irradiation.
Collapse
Affiliation(s)
- Jennifer L Spiegel
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| | - Mario Hambrecht
- Department of Otorhinolaryngology, Universitaetsmedizin Goettingen, University Medical Center Goettingen, Goettingen, Germany
| | - Vera Kohlbauer
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| | - Frank Haubner
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| | - Friedrich Ihler
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany.,German Center for Vertigo and Balance Disorders, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| | - Arndt F Schilling
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - Kai O Böker
- Department of Trauma Surgery, Orthopedics and Plastic Surgery, University Medical Center Goettingen, Goettingen, Germany
| | - Ralf Dressel
- Institute of Cellular and Molecular Immunology, University Medical Center Goettingen, Goettingen, Germany
| | - Katrin Streckfuss-Bömeke
- Department of Cardiology and Pneumology, University Medical Center Goettingen, Goettingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Goettingen, Goettingen, Germany
| | - Mark Jakob
- Department of Otorhinolaryngology, Klinikum der Universitaet Muenchen, Ludwig-Maximilians-Universitaet Muenchen, Munich, Germany
| |
Collapse
|
20
|
Li J, Chen L, Chen Q, Hu D, Lin J. [Effect of granulocyte colony-stimulating factor mobilizing bone marrow mesenchymal stell cells homing to injury sites in spinal cord injury of rats]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:93-100. [PMID: 30644268 DOI: 10.7507/1002-1892.201806127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Objective To investigate the effect of granulocyte colony-stimulating factor (G-CSF) mobilizing the bone marrow mesenchymal stem cells (BMSCs) homing to the spinal cord injury sites in rats, and to evaluate the feasibility of G-CSF mobilizing the BMSCs home to the injured spinal cord. Methods Twenty-four healthy adult female Sprague Dawley rats were injected with 1 mL green fluorescence protein labeled BMSCs (GFP-BMSCs, 1×10 6 cells/mL) into tail vein at 12 hours before operation. They were randomly divided into sham operation group (group A), sham operation+G-CSF group (group B), spinal cord injury group (group C), and spinal cord injury+G-CSF group (group D), with 6 rats in each group. In groups C and D, spinal cord injury model was established by T 10 level spinal cord hemisection. In groups A and B, only laminectomy was performed without injury to the spinal cord. Groups B and D were injected with G-CSF (10 μg/kg·d) at 1 hour after operation for 3 consecutive days, and groups A and C were injected with the same amount of saline. The Basso-Beattie-Bresnahan (BBB) score was used to estimate the neurological function of rats and the expressions of tumor necrosis factor α (TNF-α) and stromal-derived factor 1 (SDF-1) were detected by ELISA method at 1, 3, 7, 14, 21, and 28 days after operation. The spinal cord samples of rats were sacrificed at 28 days after operation for immunohistochemical staining to observe the expression of cytokines, including SDF-1, brain derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), and TNF-α, and immunofluorescence staining to observe GFP-BMSCs positive cells, double-stained fluorescent yellow GFP/neuronal nuclear antigen (NeuN) positive neurons, and GFP/glial fibrillary acidic protein (GFAP) positive neurons. The number of glial cells and apoptosis were detected by TUNEL method. Results The BBB score of groups A and B had no significant change at each time point after operation. At 1 day after operation, the BBB score of groups C and D decreased to the lowest level, and then gradually increased. The BBB score of group D was significantly higher than that of group C at all time points except 1 day after operation ( P<0.05). At 3, 7, 14, 21, 28 days after operation, the levels of TNF-α and SDF-1 in groups C and D were significantly higher than those in groups A and B ( P<0.05), but the levels of TNF-α in group D were significantly lower than those in group C at each time point, and the levels of SDF-1 were significantly higher than those in group C ( P<0.05). Immunohistochemical staining showed that the expressions of SDF-1, BDNF, VEGF, and TNF-α in groups C and D were significantly higher than those in groups A and B ( P<0.05); the expressions of SDF-1, BDNF, and VEGF in group D were significantly higher than those in group C, and the expression of TNF-α was significantly lower than that in group C ( P<0.05). Immunofluorescence staining showed that the number of GFP-BMSCs, GFP/NeuN, and GFP/GFAP positive cells in groups C and D were significantly higher than those in groups A and B, and in group D than in group C ( P<0.05). TUNEL assay showed that the number of apoptotic cells in groups C and D was significantly lower than that in groups A and B, and in group D than in group C ( P<0.05). Conclusion G-CSF can mobilize BMSCs to the spinal cord injury site and promote repair effect by down-regulating TNF-α to promote the anti-apoptosis function and up-regulating SDF-1, BDNF, VEGF to promote BMSCs migration.
Collapse
Affiliation(s)
- Jie Li
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou Fujian, 350004, P.R.China
| | - Lei Chen
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou Fujian, 350004,
| | - Qiuhong Chen
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou Fujian, 350004, P.R.China
| | - Deqing Hu
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou Fujian, 350004, P.R.China
| | - Jianhua Lin
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou Fujian, 350004, P.R.China
| |
Collapse
|
21
|
Shao Y, Zhou F, He D, Zhang L, Shen J. Overexpression of CXCR7 promotes mesenchymal stem cells to repair phosgene-induced acute lung injury in rats. Biomed Pharmacother 2018; 109:1233-1239. [PMID: 30551373 DOI: 10.1016/j.biopha.2018.10.108] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/11/2018] [Accepted: 10/20/2018] [Indexed: 12/14/2022] Open
Abstract
Phosgene exposure may result in acute lung injury (ALI) with high mortality. Emerging evidence suggests that mesenchymal stem cells (MSCs) have a therapeutic potential against ALI. CXC chemokine receptor 7 (CXCR7) has been identified as a receptor of stromal-cell-derived factor 1 (SDF1) involved in MSC migration and may be an important mediator of the therapeutic effects of MSCs on ALI. In our study, we initially constructed a lentiviral vector overexpressing CXCR7 and then successfully transduced it into rat bone marrow-derived MSCs (resulting in MSCs-CXCR7). We found that ALI and the wet-to-dry ratio significantly decreased in the phosgene-exposed rats after administration of MSCs-CXCR7 or MSCs-GFP. Indeed, treatment with MSCs-CXCR7 caused further improvement. Moreover, injection of MSCs-CXCR7 significantly facilitated MSC homing to injured lung tissue. Meanwhile, overexpression of CXCR7 promoted differentiation of MSCs into type II alveolar epithelial (AT II) cells and enhanced the ability of MSCs to modulate the inflammatory response in phosgene-induced ALI. Taken together, our findings suggest that CXCR7-overexpressing MSCs may markedly facilitate treatment of phosgene-induced ALI (P-ALI) in rats.
Collapse
Affiliation(s)
- Yiru Shao
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Fangqing Zhou
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Daikun He
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jie Shen
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China.
| |
Collapse
|
22
|
Huang S, Ren Y, Wang X, Lazar L, Ma S, Weng G, Zhao J. Application of Ultrasound-Targeted Microbubble Destruction-Mediated Exogenous Gene Transfer in Treating Various Renal Diseases. Hum Gene Ther 2018; 30:127-138. [PMID: 30205715 DOI: 10.1089/hum.2018.070] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic renal disease or acute renal injury could result in end-stage renal disease or renal failure. Sonoporation, induced by ultrasound-targeted microbubble destruction (UTMD), has evolved as a new technology for gene delivery. It increases the transfection efficiency of the genes into target kidney tissues. Moreover, UTMD-mediated gene delivery can directly repair the damaged tissues or improve the recruitment and homing of stem cells in the recovery of injured tissues, which has the potential to act as a non-viral and effective method to current gene therapy. This article reviews the mechanisms and applications of UTMD in terms of renal disease, including diabetic nephropathy, renal carcinoma, acute kidney injury, renal interstitial fibrosis, nephrotoxic nephritis, urinary stones, and acute rejection.
Collapse
Affiliation(s)
- Shuaishuai Huang
- 1 Urology and Nephrology Institute of Ningbo University, Ningbo Urology and Nephrology Hospital, Ningbo, P.R. China
| | - Yu Ren
- 1 Urology and Nephrology Institute of Ningbo University, Ningbo Urology and Nephrology Hospital, Ningbo, P.R. China
| | - Xue Wang
- 1 Urology and Nephrology Institute of Ningbo University, Ningbo Urology and Nephrology Hospital, Ningbo, P.R. China
| | - Lissy Lazar
- 2 Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, P.R. China
| | - Suya Ma
- 1 Urology and Nephrology Institute of Ningbo University, Ningbo Urology and Nephrology Hospital, Ningbo, P.R. China
| | - Guobin Weng
- 1 Urology and Nephrology Institute of Ningbo University, Ningbo Urology and Nephrology Hospital, Ningbo, P.R. China
| | - Jinshun Zhao
- 2 Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathological and Physiological Technology, Medicine School of Ningbo University, Ningbo, P.R. China
| |
Collapse
|
23
|
Hsu FT, Wei ZH, Hsuan YCY, Lin W, Su YC, Liao CH, Hsieh CL. MRI tracking of polyethylene glycol-coated superparamagnetic iron oxide-labelled placenta-derived mesenchymal stem cells toward glioblastoma stem-like cells in a mouse model. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S448-S459. [PMID: 30198338 DOI: 10.1080/21691401.2018.1499661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mesenchymal stem cells (MSCs) that display homing and infiltration properties towards tumor cells are a promising cellular targeting vector for brain tumor therapy but are limited to local-regional delivery in current preclinical models. Here, we investigated whether placenta-derived MSCs (P-MSCs) are a superior cellular vector for systemic targeting of glioblastoma stem-like cells (GSCs), with an imaging modality to real-time monitor the trafficking P-MSCs to glioblastoma sites. Results demonstrated that P-MSCs had greater migratory activity towards GSCs and across blood-brain barrier compared with bone marrow-derived MSCs, and this activity was enhanced by hypoxia precondition. Chemokine ligand 5 was identified as a chemoattractant responsible for the glioblastoma tropism of P-MSCs. Polyethylene glycol-coated superparamagnetic iron oxide (PEG-SPIO) was synthesized for cellular labelling and imaging P-MSCs, displaying high cellular uptake and no cytotoxic effect on P-MSCs cell proliferation or stemness property. The homing effects of intravenously administered PEG-SPIO-labelled P-MSCs towards intracerebral GSCs were able to be detected in mice models through T2-weighted magnetic resonance imaging (MRI). This study suggests the possibility of innovative systemic P-MSC-based cell therapy for aggressive GSCs, developing a state-of-the-art theranostic technique for real-time tracking of therapeutic P-MSCs tumor infiltration through cellular MRI.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- a Department of Radiology , School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan.,b Department of Biological Science and Technology , China Medical University , Taichung , Taiwan.,c Department of Medical Imaging , Taipei Medical University Hospital , Taipei , Taiwan.,d Research Center of Translational Imaging , College of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Zung-Hang Wei
- e Department of Power Mechanical Engineering , National Tsing Hua University , Hsinchu , Taiwan
| | | | - Willie Lin
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Yu-Chin Su
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Chia-Hui Liao
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan
| | - Chia-Ling Hsieh
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan.,h Clinical Research Center , Taipei Medical University Hospital, Taipei Medical University , Taipei , Taiwan.,i TMU Research Center of Cancer Translational Medicine , Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
24
|
Sharma A, Rani R. Do we really need to differentiate mesenchymal stem cells into insulin-producing cells for attenuation of the autoimmune responses in type 1 diabetes: immunoprophylactic effects of precursors to insulin-producing cells. Stem Cell Res Ther 2017; 8:167. [PMID: 28701182 PMCID: PMC5508489 DOI: 10.1186/s13287-017-0615-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 05/16/2017] [Accepted: 06/20/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is a multifactorial autoimmune disorder where pancreatic beta cells are lost before the clinical manifestations of the disease. Administration of mesenchymal stem cells (MSCs) or MSCs differentiated into insulin-producing cells (IPCs) have yielded limited success when used therapeutically. We have evaluated the immunoprophylactic potentials of precursors to insulin-producing cells (pIPCs) and IPCs in nonobese diabetic (NOD) mice to ask a basic question: do we need to differentiate MSCs into IPCs or will pIPCs suffice to attenuate autoimmune responses in T1D? METHODS Bone marrow-derived MSCs from Balb/c mice were characterized following the International Society for Cellular Therapy (ISCT) guidelines. MSCs cultured in high-glucose media for 11 to 13 passages were characterized for the expression of pancreatic lineage genes using real-time polymerase chain reaction. Expression of the PDX1 gene in pIPCs was assessed using Western blot and fluorescence-activated cell sorting (FACS). Triple-positive MSCs were differentiated into IPCs using a three-step protocol after sorting them for cell surface markers, i.e. CD29, CD44, and SCA-1. Nonobese diabetic mice were administered pIPCs, IPCs, or phosphate-buffered saline (PBS) into the tail vein at weeks 9 or 10 and followed-up for 29-30 weeks for fasting blood glucose levels. Two consecutive blood sugar levels of more than 250 mg/dl were considered diabetic. RESULTS MSCs grown in high-glucose media for 11 to 13 passages expressed genes of the pancreatic lineage such as PDX1, beta2, neurogenin, PAX4, Insulin, and glucagon. Furthermore, Western blot and FACS analysis for PDX-1, a transcription factor necessary for beta cell maturation, confirmed that these cells were precursors of insulin-producing cells (pIPCs). NOD mice administered with pIPCs were better protected from developing diabetes with a protective efficacy of 78.4% (p < 0.009); however, administration of IPCs gave protective efficacy of 55% at the end of 28-30 weeks. CONCLUSIONS Precursors to insulin-producing cells seem to have better potential to arrest autoimmune response in type 1 diabetes when administered before the onset of the disease in NOD mice. When translated to humans, autologous mesenchymal stem cells grown in high-glucose media for 10 to 13 passages may have beneficial effects in individuals at high risk of developing type 1 diabetes.
Collapse
Affiliation(s)
- Anshu Sharma
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, 110067, India
| | - Rajni Rani
- Molecular Immunogenetics Group, National Institute of Immunology, New Delhi, 110067, India. .,Systems Biology Group, CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110025, India.
| |
Collapse
|
25
|
Nitzsche F, Müller C, Lukomska B, Jolkkonen J, Deten A, Boltze J. Concise Review: MSC Adhesion Cascade-Insights into Homing and Transendothelial Migration. Stem Cells 2017; 35:1446-1460. [DOI: 10.1002/stem.2614] [Citation(s) in RCA: 207] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Franziska Nitzsche
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology, McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Claudia Müller
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Barbara Lukomska
- NeuroRepair Department; Mossakowski Medical Research Centre; Warsaw Poland
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern; Kuopio Finland
| | - Alexander Deten
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
| | - Johannes Boltze
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| |
Collapse
|
26
|
Mesenchymal Stem Cell Therapy for Inflammatory Skin Diseases: Clinical Potential and Mode of Action. Int J Mol Sci 2017; 18:ijms18020244. [PMID: 28125063 PMCID: PMC5343781 DOI: 10.3390/ijms18020244] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/15/2017] [Accepted: 01/18/2017] [Indexed: 02/06/2023] Open
Abstract
Inflammatory skin disorders that cause serious deterioration of the quality of life have become one of the major public concerns. Despite their significance, there is no fundamental cure to date. Mesenchymal stem cells (MSCs) possess unique immunomodulatory properties which make them a promising tool for the treatment of various inflammatory diseases. Our recent preclinical and clinical studies have shown that MSCs can be successfully used for the treatment of atopic dermatitis (AD), one of the major inflammatory skin diseases. This observation along with similar reports from other groups revealed the efficacy and underlying mechanisms of MSCs in inflammatory dermatosis. In addition, it has been proposed that cell priming or gene transduction can be novel strategies for the development of next-generation high-efficacy MSCs for treating inflammatory skin diseases. We discuss here existing evidence that demonstrates the regulatory properties of MSCs on immune responses under inflammatory conditions.
Collapse
|
27
|
A Soliman N, Abd-Allah SH, Hussein S, Alaa Eldeen M. Factors enhancing the migration and the homing of mesenchymal stem cells in experimentally induced cardiotoxicity in rats. IUBMB Life 2017; 69:162-169. [PMID: 28083978 DOI: 10.1002/iub.1600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 12/21/2016] [Indexed: 12/13/2022]
Abstract
Doxorubicin is an effective anti-neoplastic drug but its use is limited by its cardiotoxicity. Administration of mesenchymal stem cells (MSCs) for the management of cardiotoxicity was with poor myocardial homing capacity. With the aim of developing novel techniques to improve the migration of MSCs, we tested whether valproate and electric fields (EFs) direct the migration of MSCs towards the damaged myocardium. The study included five groups of female albino rats. The first group included 10 healthy rats as normal control group. The remaining 40 female rats received doxorubicin for induction of acute cardiotoxicity. Four rats were sacrificed for histopathological confirmation of cardiotoxicity. The remaining rats were equally divided into subsequent four groups. The second group included nine rats that did not receive further treatment (positive control group). The third group included nine rats which received intravenous bone marrow derived mesenchymal stem cells (BM-MSCs) after cardiotoxicity induction. The fourth group included nine rats which received BM-MSCs plus sodium valporate after cardiotoxicity induction. The fifth group included nine rats which received BM-MSCs plus sodium valporate after cardiotoxicity induction and were exposed to an electrical stimulation (ES). Blood samples were taken from all groups at the end of the study to estimate creatine kinase-MB (CK-MB), aspartate transaminase (AST) and lactate dehydrogenase (LDH). Heart tissues from all rats were used for RNA extraction for assessment of sry gene expression. Homing was tested by PKH26 fluorescence in myocardial tissue sections and by sry gene expression. The best biochemical and histopathological improvement in cardiotoxicity was demonstrated in group 5 (rats that received ES and valporate with MSCs). We concluded that EFs and sodium valproate enhance homing ability of MSCs towards the damaged myocardium in doxorubicin induced carditoxicity model. © 2017 IUBMB Life, 69(3):162-169, 2017.
Collapse
Affiliation(s)
- Nabil A Soliman
- Zoology Department, Physiology Section, Faculty of Science, Zagazig University, Egypt
| | - Somia H Abd-Allah
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Samia Hussein
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Egypt
| | - Muhammad Alaa Eldeen
- Zoology Department, Physiology Section, Faculty of Science, Zagazig University, Egypt
| |
Collapse
|
28
|
Wang G, Zhang Q, Zhuo Z, Wu S, Xu Y, Zou L, Gan L, Tan K, Xia H, Liu Z, Gao Y. Enhanced Homing of CXCR-4 Modified Bone Marrow-Derived Mesenchymal Stem Cells to Acute Kidney Injury Tissues by Micro-Bubble-Mediated Ultrasound Exposure. ULTRASOUND IN MEDICINE & BIOLOGY 2016; 42:539-548. [PMID: 26610714 DOI: 10.1016/j.ultrasmedbio.2015.10.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 10/04/2015] [Accepted: 10/10/2015] [Indexed: 06/05/2023]
Abstract
Although the curative effects of bone marrow stromal cells (BMSCs) for acute kidney injury (AKI) have been recognized, their in vivo reparative capability is limited by the low levels of targeted homing and retention of intravenous injected cells. Stromal cell-derived factor-1 (SDF-1) plays an important role in stem cell homing and retention through interaction with its specific functional receptor, CXCR4, which is presumably related to the poor homing in AKI therapy. However, most of the functional CXCR4 chemokine receptors are lost upon in vitro culturing. Ultrasound-targeted micro-bubble destruction (UTMD) has become one of the most promising strategies for the targeted delivery of drugs and genes. To improve BMSC homing to AKI kidneys, we isolated and cultured rat BMSCs to third passage and enhanced CXCR-4 transfection efficiency in vitro by applying UTMD and polyethylenimine. Transwell migration assay showed that the migration ability of CXCR4-modified BMSCs was nine-fold higher than controls. Then, mercuric chloride-induced AKI rats were injected with transfected BMSCs through their tail veins. We showed that enhanced homing and retention of BMSCs were observed in the CXCR-4 modified group compared with other groups at 1, 2 and 3 d post-treatment. Collectively, our data indicated that UTMD was an effective method to increase BMSCs' engraftment to AKI kidney tissues by increasing CXCR-4 expression.
Collapse
Affiliation(s)
- Gong Wang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Qian Zhang
- Department of Nephropathy, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongxiong Zhuo
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Shengzheng Wu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Linru Zou
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Ling Gan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Kaibin Tan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Hongmei Xia
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yunhua Gao
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
29
|
Gao F, Chiu SM, Motan DAL, Zhang Z, Chen L, Ji HL, Tse HF, Fu QL, Lian Q. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis 2016; 7:e2062. [PMID: 26794657 PMCID: PMC4816164 DOI: 10.1038/cddis.2015.327] [Citation(s) in RCA: 768] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/13/2015] [Accepted: 09/25/2015] [Indexed: 12/11/2022]
Abstract
The unique immunomodulatory properties of mesenchymal stem cells (MSCs) make them an invaluable cell type for the repair of tissue/ organ damage caused by chronic inflammation or autoimmune disorders. Although they hold great promise in the treatment of immune disorders such as graft versus host disease (GvHD) and allergic disorders, there remain many challenges to overcome before their widespread clinical application. An understanding of the biological properties of MSCs will clarify the mechanisms of MSC-based transplantation for immunomodulation. In this review, we summarize the preclinical and clinical studies of MSCs from different adult tissues, discuss the current hurdles to their use and propose the future development of pluripotent stem cell-derived MSCs as an approach to immunomodulation therapy.
Collapse
Affiliation(s)
- F Gao
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - S M Chiu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - D A L Motan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Z Zhang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - L Chen
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - H-L Ji
- Department of Cellular and Molecular Biology, University of Texas Health Science Center at Tyler, Tyler, Texas 75708, USA
| | - H-F Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Q-L Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Q Lian
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| |
Collapse
|
30
|
Effects of diagnostic ultrasound-targeted microbubble destruction on the homing ability of bone marrow stromal cells to the kidney parenchyma. Eur Radiol 2015; 26:3006-16. [DOI: 10.1007/s00330-015-4123-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/24/2015] [Accepted: 11/16/2015] [Indexed: 12/13/2022]
|
31
|
Zhou Y, Hu Q, Chen F, Zhang J, Guo J, Wang H, Gu J, Ma L, Ho G. Human umbilical cord matrix-derived stem cells exert trophic effects on β-cell survival in diabetic rats and isolated islets. Dis Model Mech 2015; 8:1625-33. [PMID: 26398949 PMCID: PMC4728317 DOI: 10.1242/dmm.021857] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/11/2015] [Indexed: 02/05/2023] Open
Abstract
Human umbilical cord matrix-derived stem cells (uMSCs), owing to their cellular and procurement advantages compared with mesenchymal stem cells derived from other tissue sources, are in clinical trials to treat type 1 (T1D) and type 2 diabetes (T2D). However, the therapeutic basis remains to be fully understood. The immunomodulatory property of uMSCs could explain the use in treating T1D; however, the mere immune modulation might not be sufficient to support the use in T2D. We thus tested whether uMSCs could exert direct trophic effects on β-cells. Infusion of uMSCs into chemically induced diabetic rats prevented hyperglycemic progression with a parallel preservation of islet size and cellularity, demonstrating the protective effect of uMSCs on β-cells. Mechanistic analyses revealed that uMSCs engrafted long-term in the injured pancreas and the engraftment markedly activated the pancreatic PI3K pathway and its downstream anti-apoptotic machinery. The pro-survival pathway activation was associated with the expression and secretion of β-cell growth factors by uMSCs, among which insulin-like growth factor 1 (IGF1) was highly abundant. To establish the causal relationship between the uMSC-secreted factors and β-cell survival, isolated rat islets were co-cultured with uMSCs in the transwell system. Co-culturing improved the islet viability and insulin secretion. Furthermore, reduction of uMSC-secreted IGF1 via siRNA knockdown diminished the protective effects on islets in the co-culture. Thus, our data support a model whereby uMSCs exert trophic effects on islets by secreting β-cell growth factors such as IGF1. The study reveals a novel therapeutic role of uMSCs and suggests that multiple mechanisms are employed by uMSCs to treat diabetes.
Collapse
Affiliation(s)
- Yunting Zhou
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Qi Hu
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Fuyi Chen
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Juan Zhang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jincheng Guo
- Department of Molecular Pathology, Shantou University Medical College, Shantou 515041, China
| | - Hongwu Wang
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jiang Gu
- Department of Molecular Pathology, Shantou University Medical College, Shantou 515041, China
| | - Lian Ma
- Department of Pediatrics, The Women and Children's Hospital of Shenzhen University, Shenzhen 518122, China
| | - Guyu Ho
- Department of Pediatrics, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
32
|
Wang G, Zhuo Z, Yang B, Wu S, Xu Y, Liu Z, Tan K, Xia H, Wang X, Zou L, Gan L, Gao Y. Enhanced Homing Ability and Retention of Bone Marrow Stromal Cells to Diabetic Nephropathy by Microbubble-Mediated Diagnostic Ultrasound Irradiation. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2977-2989. [PMID: 26318561 DOI: 10.1016/j.ultrasmedbio.2015.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/22/2015] [Accepted: 07/06/2015] [Indexed: 06/04/2023]
Abstract
Bone marrow stromal cell (BMSC) transplantation can successfully treat diabetic nephropathy (DN), but the lack of a specific homing place for intravenously injected cells limits the effective implementation of stem cell therapies. The migration and survival of transplanted BMSCs are determined by inflammatory reactions in the local kidney micro-environment. We tested the hypothesis that microbubble-mediated diagnostic ultrasound irradiation could provide a suitable micro-environment for BMSC delivery and retention in DN therapy. In this study, red fluorescent protein-labeled BMSCs were administered combined with microbubbles to streptozotocin-induced DN rats 4 wk after diabetes onset. We observed enhanced BMSC homing and retention in microbubble-mediated diagnostic ultrasound-irradiated kidneys compared with the contralateral kidneys on days 1 and 3 post-treatment. The results from immunohistochemical analysis, Western blot and enzyme-linked immunosorbent assay indicated that the local and transient expression of various chemo-attractants (i.e., cytokines, integrins and trophic factors) found to promote BMSC homing was much higher than observed in non-treated kidneys. The local capillary endothelium rupture observed by transmission electron microscopy may account for local micro-environment changes. Histopathologic analysis revealed no signs of kidney damage. These results confirmed that renal micro-environment changes caused by appropriate microbubble-mediated diagnostic ultrasound irradiation may promote BMSC homing ability to the diabetic kidney without renal toxicity and cell damage. This non-invasive and effective technique may be a promising method for BMSC transplantation therapy.
Collapse
Affiliation(s)
- Gong Wang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongxiong Zhuo
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Bin Yang
- Department of Ultrasound Diagnostics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Shengzheng Wu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Kaibin Tan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Hongmei Xia
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoyan Wang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Linru Zou
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Ling Gan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yunhua Gao
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
33
|
Cerri S, Greco R, Levandis G, Ghezzi C, Mangione AS, Fuzzati-Armentero MT, Bonizzi A, Avanzini MA, Maccario R, Blandini F. Intracarotid Infusion of Mesenchymal Stem Cells in an Animal Model of Parkinson's Disease, Focusing on Cell Distribution and Neuroprotective and Behavioral Effects. Stem Cells Transl Med 2015. [PMID: 26198165 DOI: 10.5966/sctm.2015-0023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
UNLABELLED Mesenchymal stem cells (MSCs) have been proposed as a potential therapeutic tool for Parkinson's disease (PD) and systemic administration of these cells has been tested in preclinical and clinical studies. However, no information on survival and actual capacity of MSCs to reach the brain has been provided. In this study, we evaluated homing of intraarterially infused rat MSCs (rMSCs) in the brain of rats bearing a 6-hydroxydopamine (6-OHDA)-induced lesion of the nigrostriatal tract, to establish whether the toxin-induced damage is sufficient to grant MSC passage across the blood-brain barrier (BBB) or if a transient BBB disruption is necessary. The rMSC distribution in peripheral organs and the effects of cell infusion on neurodegenerative process and motor deficits were also investigated. rMSCs were infused 14 days after 6-OHDA injection. A hyperosmolar solution of mannitol was used to transiently permeabilize the BBB. Behavioral impairment was assessed by adjusting step test and response to apomorphine. Animals were sacrificed 7 and 28 days after cell infusion. Our work shows that appreciable delivery of rMSCs to the brain of 6-OHDA-lesioned animals can be obtained only after mannitol pretreatment. A notable percentage of infused cells accumulated in peripheral organs. Infusion of rMSCs did not modify the progression of 6-OHDA-induced damage or the motor impairment at the stepping test, but induced progressive normalization of the pathological response (contralateral turning) to apomorphine administration. These findings suggest that many aspects should be further investigated before considering any translation of MSC systemic administration into the clinical setting for PD treatment. SIGNIFICANCE This study demonstrates that mesenchymal stem cells infused through the carotid artery do not efficiently cross the blood-brain barrier in rats with a Parkinson's disease-like degeneration of nigrostriatal neurons, unless a permeabilizing agent (e.g., mannitol) is used. The infusion did not reduce the neuronal damage and associated motor impairment, but abolished the motor abnormalities these animals typically show when challenged with a dopaminergic agonist. Therefore, although arterially infused mesenchymal stem cells did not show neurorestorative effects in this study's Parkinson's disease model, they appeared to normalize the pathological responsiveness of striatal neurons to dopaminergic stimulation. This capability should be further explored in future studies.
Collapse
Affiliation(s)
- Silvia Cerri
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rosaria Greco
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giovanna Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Cristina Ghezzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Antonina Stefania Mangione
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marie-Therese Fuzzati-Armentero
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arianna Bonizzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Antonietta Avanzini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rita Maccario
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, and Laboratory of Neurophysiology of Integrative Autonomic Systems, "C. Mondino" National Neurological Institute, Pavia, Italy; Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
34
|
Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant 2015; 23:1045-59. [PMID: 23676629 DOI: 10.3727/096368913x667709] [Citation(s) in RCA: 639] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are one of a few stem cell types to be applied in clinical practice as therapeutic agents for immunomodulation and ischemic tissue repair. In addition to their multipotent differentiation potential, a strong paracrine capacity has been proposed as the principal mechanism that contributes to tissue repair. Apart from cytokine/chemokine secretion, MSCs also display a strong capacity for mitochondrial transfer and microvesicle (exosomes) secretion in response to injury with subsequent promotion of tissue regeneration. These unique properties of MSCs make them an invaluable cell type to repair damaged tissues/organs. Although MSCs offer great promise in the treatment of degenerative diseases and inflammatory disorders, there are still many challenges to overcome prior to their widespread clinical application. Particularly, their in-depth paracrine mechanisms remain a matter for debate and exploration. This review will highlight the discovery of the paracrine mechanism of MSCs, regulation of the paracrine biology of MSCs, important paracrine factors of MSCs in modulation of tissue repair, exosome and mitochondrial transfer for tissue repair, and the future perspective for MSC-based therapy.
Collapse
Affiliation(s)
- Xiaoting Liang
- Cardiology Division, Department of Medicine, University of Hong Kong, Hong Kong
| | | | | | | | | |
Collapse
|
35
|
Golzar F, Javanmard SH, Bahrambeigi V, Rafiee L. The effect of Kisspeptin-10 on mesenchymal stem cells migration in vitro and in vivo. Adv Biomed Res 2015; 4:20. [PMID: 25709985 PMCID: PMC4333426 DOI: 10.4103/2277-9175.149851] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 12/14/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Kisspeptins (kp) activate a receptor coupled to a Gαq subunit (GPR54 or KiSS-1R) receptor to perform a variety of functions, including inhibition of cell motility, chemotaxis, and metastasis. In this study we have investigated whether kp-10, the most potent member of the kisspeptin family, can modulate CXCR4 (C-X-C chemokine receptor type 4) expression and mesenchymal stem cells (MSCs) migration that may influence the development of tumors. MATERIALS AND METHODS We compared the directional migration of MSCs treated with 10-100 or 500 nM kp-10 for 24 hours and no treated cells using an in vitro transmembrane migration assay. In addition, Chloromethylbenzamido Dialkylacarbocyanine (CM-Dil) labeled adipose-derived mesenchymal stem cells treated with 10-100 or 500 nM kp-10 and no treated cells were transfused via the tail vein to the melanoma tumor bearing C57BL/6 mice. After 24 hours, the mice were scarified, the tumors were dissected, and the tumor cell suspensions were analyzed by flow cytometry for detection of CM-Dil(+) MSCs. RESULTS We have found that kp-10 increased the MSCs migration at 100 nM, while it decreased the MSCs migration at 500 nM, both in vitro and in vivo, with a significant increase of CXCR4 expression at 100 nM kp-10 compared to the no treated cells, but it had no significant difference between the various concentrations of kp-10. CONCLUSION Thus, our data showed that kp-10 can differently affect MSCs migration in various concentrations, probably through different effects on CXCR4 expression in various concentrations.
Collapse
Affiliation(s)
- Fatemeh Golzar
- Student's Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran ; Physiology Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Vahid Bahrambeigi
- Physiology Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Laleh Rafiee
- Physiology Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Bone marrow stromal cells as immunomodulators. A primer for dermatologists. J Dermatol Sci 2014; 77:11-20. [PMID: 25476233 DOI: 10.1016/j.jdermsci.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023]
Abstract
Bone marrow stromal cells (BMSCs, also known as mesenchymal stem cells or MSCs) represent a unique cell population in the bone marrow with a long-known function to support hematopoiesis and replace skeletal tissues. The recent discovery that BMSCs also possess potent immunoregulatory features attracted a great deal of attention from stem cell biologists, immunologists and clinicians of different specialties worldwide. Initial clinical experience along with several animal models suggested that intravenously delivered BMSCs are able to regulate a wide variety of host immune cells and act in a way that is beneficial for the recipient in a variety of diseases. The role of the present review is to give a short introduction to the biology of BMSCs and to summarize our current understanding of how BMSCs modulate the immune system with special emphasis on available clinical data. Considering the audience of this journal we will also attempt to guide dermatologists in choosing the right skin conditions where BMSCs might be considered as a therapeutic alternative.
Collapse
|
37
|
|
38
|
Wyse RD, Dunbar GL, Rossignol J. Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases. Int J Mol Sci 2014; 15:1719-45. [PMID: 24463293 PMCID: PMC3958818 DOI: 10.3390/ijms15021719] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/01/2023] Open
Abstract
The transplantation of mesenchymal stem cells (MSCs) for treating neurodegenerative disorders has received growing attention recently because these cells are readily available, easily expanded in culture, and when transplanted, survive for relatively long periods of time. Given that such transplants have been shown to be safe in a variety of applications, in addition to recent findings that MSCs have useful immunomodulatory and chemotactic properties, the use of these cells as vehicles for delivering or producing beneficial proteins for therapeutic purposes has been the focus of several labs. In our lab, the use of genetic modified MSCs to release neurotrophic factors for the treatment of neurodegenerative diseases is of particular interest. Specifically, glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain derived neurotrophic factor (BDNF) have been recognized as therapeutic trophic factors for Parkinson's, Alzheimer's and Huntington's diseases, respectively. The aim of this literature review is to provide insights into: (1) the inherent properties of MSCs as a platform for neurotrophic factor delivery; (2) the molecular tools available for genetic manipulation of MSCs; (3) the rationale for utilizing various neurotrophic factors for particular neurodegenerative diseases; and (4) the clinical challenges of utilizing genetically modified MSCs.
Collapse
Affiliation(s)
- Robert D Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| |
Collapse
|
39
|
Zhang D, Zheng L, Shi H, Chen X, Wan Y, Zhang H, Li M, Lu L, Luo S, Yin T, Lin H, He S, Luo Y, Yang L. Suppression of peritoneal tumorigenesis by placenta-derived mesenchymal stem cells expressing endostatin on colorectal cancer. Int J Med Sci 2014; 11:870-9. [PMID: 25013366 PMCID: PMC4081308 DOI: 10.7150/ijms.8758] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/11/2014] [Indexed: 02/05/2023] Open
Abstract
MSCs-based therapy for cancer is a relatively new but rapidly growing area of research. Human term placenta, an attractive source of MSCs (PMSCs), appears to have great advantage due to its easy access without invasive procedures, its lack of ethical issues and its high-throughput and young age. In the present study, we isolated MSCs from placenta and characterized their morphology and differentiation capacities. We next investigated the underlying antitumor effects and the potential mechanism of PMSCs to express endostatin using adenoviral transduction (Ad-Endo) in a colorectal peritoneal carcinomatosis (CRPC) mouse model. For in vitro experiments, the migratory potential of Ad-Endo-PMSCs towards tumor cells was demonstrated using a double-chamber assay, and the anti-angiogenesis ability of endostatin from engineered PMSCs was evaluated using the tube formation assay. For the in vivo study, mice harboring CT26 colorectal cancer indicated a significant reduction in tumor nodules and a prolongation of survival following Ad-Endo-PMSCs therapy. These observations were associated with significantly decreased tumor cell proliferation and blood vessel counts as well as increased tumor cell apoptosis. These data suggested the potential of PMSCs-based gene therapy for the targeted delivery of therapeutic proteins in cancer.
Collapse
Affiliation(s)
- Dongmei Zhang
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Lan Zheng
- 2. Department of Obstetrics and Gynecology, Second West China Hospital of Sichuan University, Chengdu, 610041, P. R. China
| | - Huashan Shi
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiancheng Chen
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yang Wan
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Hailong Zhang
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Meng Li
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Lian Lu
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Shuntao Luo
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Tao Yin
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Honggang Lin
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Shasha He
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Yan Luo
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Li Yang
- 1. State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
40
|
Abstract
Pericytes, typically attached to the walls of microvessels in almost all organs, interact with endothelial cells and take part in diverse biological processes, e.g. blood vessel regulation and tissue repair. This suggests that pericytes harbor a remarkable degree of cellular plasticity, which could potentially be employed for the treatment of diseases affecting diverse tissues such as the skeletal muscle and the central nervous system. Here, we follow pericytes on their journey across Waddington's epigenetic landscape, descending from their origin, along a path guided by environmental signals or ectopic transcription factors, at the end of which they acquire a new identity, e.g. muscle or nerve cells. The central theme of this review is the question of whether pericytes can be enticed to differentiate into whatever cell type is needed, and thus provide an endogenous cellular source for treating as yet incurable diseases--like a magic bullet.
Collapse
Affiliation(s)
- Marisa Karow
- Department of Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany.
| |
Collapse
|
41
|
The use of hypoxic cultured mesenchymal stem cell for oncolytic virus therapy. Cancer Gene Ther 2013; 20:308-16. [PMID: 23618949 DOI: 10.1038/cgt.2013.22] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The safety of oncolytic viruses, such as conditionally replicative adenoviruses (CRAds), has been validated in clinical trials for cancer therapy. Their antitumor efficacy is limited by the presence of preexisting neutralizing antibodies (NAbs). Mesenchymal stem cells (MSCs) are attractive as a cellular vehicle to carry antitumor agents, not only because they are easily obtained and expanded to great numbers in vitro, but also because of their ability to migrate and engraft to tumors. MSCs expanded under hypoxic conditions decrease in replicative senescence and increase in proliferation capacity and differentiation potentials. However it remains to be clarified whether these hypoxic MSCs also are good carriers for the delivery of CRAds to tumor cells in the presence of NAbs. This study firstly demonstrated hypoxic MSCs with an increased ability to migrate toward tumors through the upregulation of chemokine receptors, such as CXCR4 and CX3CR1. It is then demonstrated that hypoxic MSCs has the capacity to carry CRAds, without inducing apoptosis, for up to one week. Using an in vitro coculture with human colon cancer cells and with intraperitoneally (i.p.) and subcutaneously (s.c.) developed human colon cancer xenografts, it is demonstrated that hypoxic MSCs are able to protect CRAds from attack by NAbs, thereby successfully delivering them to the target tumor cells. These results show that hypoxic MSCs can serve as cell carriers for CRAds and may help to develop new strategies against cancer.
Collapse
|
42
|
Yulyana Y, Endaya BB, Ng WH, Guo CM, Hui KM, Lam PYP, Ho IAW. Carbenoxolone enhances TRAIL-induced apoptosis through the upregulation of death receptor 5 and inhibition of gap junction intercellular communication in human glioma. Stem Cells Dev 2013; 22:1870-82. [PMID: 23428290 DOI: 10.1089/scd.2012.0529] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been used extensively in cancer therapy. However, more than half of glioblastoma multiforme are insensitive to the apoptotic effect of TRAIL. Improvement in therapeutic modalities that enhances the efficacy of TRAIL in glioma is much sought after. In this study, we combined the tumor selectivity of TRAIL and tumor-homing properties of mesenchymal stem cells (MSC) with gap junction (GJ) inhibitory effect of carbenoxolone (CBX) to target orthotopic glioma. MSC were engineered to express TRAIL (MSC-TRAIL) by incorporating the secretable trimeric form of TRAIL into a Herpes Simplex Virus (HSV) type I amplicon vector. Our results showed that combined treatment of MSC-TRAIL and CBX enhanced glioma cell death, especially in three primary human glioma isolates, of which two of those are marginally sensitive to TRAIL. CBX enhanced TRAIL-induced apoptosis through upregulation of death receptor 5, blockade of GJ intercellular communication, and downregulation of connexin 43. Dual arm therapy using TRAIL and CBX prolonged the survival of treated mice by ~27% when compared with the controls in an intracranial glioma model. The enhanced efficacy of TRAIL in combination with CBX coupled with the minimal cytotoxic nature of CBX suggested a favorable clinical usage of this treatment regimen.
Collapse
Affiliation(s)
- Yulyana Yulyana
- Laboratory of Cancer Gene Therapy, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
43
|
Dreher L, Elvers-Hornung S, Brinkmann I, Huck V, Henschler R, Gloe T, Klüter H, Bieback K. Cultivation in Human Serum Reduces Adipose Tissue-Derived Mesenchymal Stromal Cell Adhesion to Laminin and Endothelium and Reduces Capillary Entrapment. Stem Cells Dev 2013; 22:791-803. [DOI: 10.1089/scd.2012.0051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Lena Dreher
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Susanne Elvers-Hornung
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Irena Brinkmann
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Volker Huck
- Department of Dermatology, Experimental Dermatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Reinhard Henschler
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Institute of Transfusion Medicine and Immune Haematology, Frankfurt, Germany
| | - Torsten Gloe
- Cardiovascular Physiology, Medical Faculty Mannheim, Centre for Biomedicine and Medical Technology, Heidelberg University, Mannheim, Germany
| | - Harald Klüter
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| | - Karen Bieback
- German Red Cross Blood Service of Baden-Württemberg–Hessen, Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, Heidelberg University, Mannheim, Germany
| |
Collapse
|
44
|
Casado JG, Tarazona R, Sanchez-Margallo FM. NK and MSCs Crosstalk: The Sense of Immunomodulation and Their Sensitivity. Stem Cell Rev Rep 2013; 9:184-9. [DOI: 10.1007/s12015-013-9430-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
45
|
Kholodenko IV, Konieva AA, Kholodenko RV, Yarygin KN. Molecular mechanisms of migration and homing of intravenously transplanted mesenchymal stem cells. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2050-1218-2-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
46
|
Vegh I, Grau M, Gracia M, Grande J, de la Torre P, Flores AI. Decidua mesenchymal stem cells migrated toward mammary tumors in vitro and in vivo affecting tumor growth and tumor development. Cancer Gene Ther 2013; 20:8-16. [PMID: 23037810 DOI: 10.1038/cgt.2012.71] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mesenchymal stem cells (MSCs) have affinity to tumor sites where they home, affecting their biology and growth. Previously, we have isolated mesenchymal cells from the decidua of the human placenta named as decidua-derived MSCs (DMSCs). The aims of the present study were to investigate the migration capacity of DMSCs in vitro, and in vivo in a preclinical model of mammary tumors induced by N-nitroso-N-methylurea (NMU). Additionally, we assessed the safety of DMSC administration in vivo and their effect on tumor growth. In vitro studies showed that DMSCs significantly migrate toward both, healthy human breast tissue and breast adenocarcinoma. Nevertheless, the effect on DMSC migration was significantly higher in the presence of tumor tissue. DMSCs also significantly migrated in vitro in the presence of NMU-mammary tumor homogenate when compared with control media alone. In vivo studies showed both migration and engraftment of DMSCs into NMU-induced tumors. Interestingly, DMSCs showed an inhibitory effect on the growth of primary tumors and in the development of new tumors. DMSCs did not affect the growth of secondary tumors, although secondary tumors appeared 2 weeks later, and the number of secondary tumors was lower in the DMSC-treated rats as compared with vehicle-treated rats. To our knowledge, this is the first report showing placental MSCs effect on tumor growth. In conclusion, DMSCs could serve as a therapeutic agent themselves and as a cellular vehicle of anticancer drugs.
Collapse
Affiliation(s)
- I Vegh
- Cancer Biology, Research Center, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Pulmonary fibrosis is a feature of a number of important lung diseases, and alveolar epithelial injury plays a key role in their pathogenesis. Traditionally, type II alveolar epithelial cells have been viewed as the progenitor cells of the alveolar epithelium; however, recent studies have identified a number of other progenitor and stem cell populations that may participate in alveolar epithelial repair. These studies suggest that the injury microenvironment plays a role in regulation of progenitor cell populations. In human idiopathic pulmonary fibrosis, epithelial abnormalities including altered cell cycling characteristics, hyperplasia, and metaplasia are observed, suggesting that dysregulation of epithelial progenitor cells contributes to the characteristic aberrant repair process. Reactivation of developmental signaling pathways such as the Wnt-β-catenin pathway is implicated in the dysregulation of these cells, and targeting these pathways may provide opportunities for therapeutic intervention. There has been a great deal of interest in the delivery of exogenous stem cells as a therapeutic strategy, and various stem and progenitor cell populations have improved outcomes in animal lung fibrosis models. The contributions of these cells to alveolar epithelial regeneration have been variable, and secretion of soluble mediators has been implicated in the beneficial effects. It remains to be seen whether the promising results seen in the preclinical studies will translate to human disease, and the first studies using mesenchymal stem cells in clinical trials for fibrotic lung disease are underway. Strategies using other stem cell populations hold promise, but currently these are a lot further from the bedside.
Collapse
|
48
|
Ziadloo A, Burks SR, Gold EM, Lewis BK, Chaudhry A, Merino MJ, Frenkel V, Frank JA. Enhanced homing permeability and retention of bone marrow stromal cells by noninvasive pulsed focused ultrasound. Stem Cells 2012; 30:1216-27. [PMID: 22593018 DOI: 10.1002/stem.1099] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bone marrow stromal cells (BMSCs) have shown significant promise in the treatment of disease, but their therapeutic efficacy is often limited by inefficient homing of systemically administered cells, which results in low number of cells accumulating at sites of pathology. BMSC home to areas of inflammation where local expression of integrins and chemokine gradients is present. We demonstrated that nondestructive pulsed focused ultrasound (pFUS) exposures that emphasize the mechanical effects of ultrasound-tissue interactions induced local and transient elevations of chemoattractants (i.e., cytokines, integrins, and growth factors) in the murine kidney. pFUS-induced upregulation of cytokines occurred through approximately 1 day post-treatment and returned to contralateral kidney levels by day 3. This window of significant increases in cytokine expression was accompanied by local increases of other trophic factors and integrins that have been shown to promote BMSC homing. When BMSCs were intravenously administered following pFUS treatment to a single kidney, enhanced homing, permeability, and retention of BMSC was observed in the treated kidney versus the contralateral kidney. Histological analysis revealed up to eight times more BMSC in the peritubular regions of the treated kidneys on days 1 and 3 post-treatment. Furthermore, cytokine levels in pFUS-treated kidneys following BMSC administration were found to be similar to controls, suggesting modulation of cytokine levels by BMSC. pFUS could potentially improve cell-based therapies as a noninvasive modality to target homing by establishing local chemoattractant gradients and increasing expression of integrins to enhance tropism of cells toward treated tissues.
Collapse
Affiliation(s)
- Ali Ziadloo
- Laboratory of Diagnostic Radiology Research, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Wadhawan N, Kalkat H, Natarajan K, Ma X, Gajjeraman S, Nandagopal S, Hao N, Li J, Zhang M, Deng J, Xiang B, Mzengeza S, Freed DH, Arora RC, Tian G, Lin F. Growth and positioning of adipose-derived stem cells in microfluidic devices. LAB ON A CHIP 2012; 12:4829-4834. [PMID: 23007449 DOI: 10.1039/c2lc40891k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Stem cells hold great promise for treatment of various degenerative diseases. However, clinical studies have only shown very moderate benefits of cell therapy. We believe that insufficiency of therapeutic benefits is due to limited homing of implanted stem cells to targeted organs. Microfluidic devices are a very useful research tool for quantitative characterizations of stem cells. The present study therefore was to assess the effects of epidermal growth factor (EGF) and direct current electric field (dcEF) on the growth and trafficking of adipose-derived stem cells (ASC). It was found that EGF did not affect cell proliferation in cell-culture flasks. However, ASC proliferated at a higher rate in microfluidic devices with continuous infusion of EGF. Furthermore, we found that ASC migrated toward an EGF gradient in microfluidic devices. Moreover, we found that ASC tended to position perpendicularly to dcEF. The results suggest that EGF and dcEF may be effective in guiding homing and trafficking of implanted ASC.
Collapse
Affiliation(s)
- Nitin Wadhawan
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mesenchymal stem cells as therapeutic agents and potential targeted gene delivery vehicle for brain diseases. J Control Release 2012; 162:464-73. [DOI: 10.1016/j.jconrel.2012.07.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 07/23/2012] [Accepted: 07/25/2012] [Indexed: 01/01/2023]
|