1
|
Liu G, Yang C, Wang X, Chen X, Cai H, Le W. Cerebellum in neurodegenerative diseases: Advances, challenges, and prospects. iScience 2024; 27:111194. [PMID: 39555407 PMCID: PMC11567929 DOI: 10.1016/j.isci.2024.111194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a group of neurological disorders characterized by the progressive dysfunction of neurons and glial cells, leading to their structural and functional degradation in the central and/or peripheral nervous system. Historically, research on NDs has primarily focused on the brain, brain stem, or spinal cord associated with disease-related symptoms, often overlooking the role of the cerebellum. However, an increasing body of clinical and biological evidence suggests a significant connection between the cerebellum and NDs. In several NDs, cerebellar pathology and biochemical changes may start in the early disease stages. This article provides a comprehensive update on the involvement of the cerebellum in the clinical features and pathogenesis of multiple NDs, suggesting that the cerebellum is involved in the onset and progression of NDs through various mechanisms, including specific neurodegeneration, neuroinflammation, abnormal mitochondrial function, and altered metabolism. Additionally, this review highlights the significant therapeutic potential of cerebellum-related treatments for NDs.
Collapse
Affiliation(s)
- Guangdong Liu
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cui Yang
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xin Wang
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xi Chen
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weidong Le
- Institute of Neurology, Sichuan Academy of Medical Sciences-Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
- Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai 200237, China
| |
Collapse
|
2
|
Zhang C, Su K, Jiang X, Tian Y, Li K. Advances in research on potential therapeutic approaches for Niemann-Pick C1 disease. Front Pharmacol 2024; 15:1465872. [PMID: 39263569 PMCID: PMC11387184 DOI: 10.3389/fphar.2024.1465872] [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: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024] Open
Abstract
Niemann-Pick disease type C1 (NP-C1) is a rare and devastating recessive inherited lysosomal lipid and cholesterol storage disorder caused by mutations in the NPC1 or NPC2 gene. These two proteins bind to cholesterol and cooperate in endosomal cholesterol transport. Characteristic clinical manifestations of NP-C1 include hepatosplenomegaly, progressive neurodegeneration, and ataxia. While the rarity of NP-C1 presents a significant obstacle to progress, researchers have developed numerous potential therapeutic approaches over the past two decades to address this condition. Various methods have been proposed and continuously improved to slow the progression of NP-C1, although they are currently at an animal or clinical experimental stage. This overview of NP-C1 therapy will delve into different theoretical treatment strategies, such as small molecule therapies, cell-based approaches, and gene therapy, highlighting the complex therapeutic challenges associated with this disorder.
Collapse
Affiliation(s)
- Caifeng Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Keke Su
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- First College for Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xu Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- First College for Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yuping Tian
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- First College for Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ke Li
- Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- First College for Clinical Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
3
|
Yang M, Zhao Y, Li X, Li H, Cheng F, Liu Y, Jia Z, He Y, Lin J, Guan L. Conditioned medium of human menstrual blood-derived endometrial stem cells protects against cell inflammation and apoptosis of Npc1 KO N2a cells. Metab Brain Dis 2023; 38:2301-2313. [PMID: 37261632 DOI: 10.1007/s11011-023-01243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023]
Abstract
Niemann-Pick disease type C1 (NPC1) is a hereditary neurodegenerative disorder caused by a mutation in the NPC1 gene. This gene encodes a transmembrane protein found in lysosomes. This disease characterized by hepatosplenomegaly, neurological impairments and premature death. Recent preclinical studies have shown promising results in using mesenchymal stem cells (MSCs) to alleviate the symptoms of NPC1. One type of MSCs, known as human menstrual blood-derived endometrial stem cells (MenSCs), has attracted attention due to its accessibility, abundant supply, and strong proliferation and regeneration capabilities. However, it remains uncertain whether the conditioned medium of MenSCs (MenSCs-CM) can effectively relieve the symptoms of NPC1. To investigate this further, we employed the CRISPR-Cas9 technique to successfully create a Npc1 gene knockout N2a cell line (Npc1KO N2a). Sanger sequencing confirmed the occurrence of Npc1 gene mutation in these cells, while western blotting revealed a lack of NPC1 protein expression. Filipin staining provided visual evidence of unesterified cholesterol accumulation in Npc1KO N2a cells. Moreover, Npc1KO N2a cells exhibited significantly decreased viability, increased inflammation, and heightened cell apoptosis. Notably, our study demonstrated that the viability of Npc1KO N2a cells was most significantly improved after being cultured by 36 h-collected MenSCs-CM for 0.5 days. Additionally, MenSCs-CM exhibited the ability to effectively reduce inflammation, counteract cell apoptosis, and ameliorate unesterified cholesterol accumulation in Npc1KO N2a cells. This groundbreaking finding establishes, for the first time, the protective effect of MenSCs-CM on N2a cells with Npc1 gene deletion. These findings suggest that the potential of MenSCs-CM as a beneficial therapeutic approach for NPC1 and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Minlin Yang
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Yanchun Zhao
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Xiaoying Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Han Li
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Fangfang Cheng
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Yanli Liu
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Zisen Jia
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China
| | - Ya'nan He
- Zhongyuan Stem Cell Research Institute, Xinxiang, Henan, 453003, China
| | - Juntang Lin
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| | - Lihong Guan
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, 453003, China.
| |
Collapse
|
4
|
Soliman AH, Abdellatif M. COVID-19 disease treatment: pivotal challenges in the arena of umbilical cord-mesenchymal stem cells (UC-MSCs). Front Cell Dev Biol 2023; 11:1146835. [PMID: 37274737 PMCID: PMC10235792 DOI: 10.3389/fcell.2023.1146835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023] Open
Abstract
This century's first major epidemic of a new coronavirus illness (2019-nCoV) was a tremendous shock to the healthcare system. The onset of the pandemic has caused severe economic and health shortages. At this time, there are no viable treatments for COVID-19. Several clinical studies using cell-based therapies, such as umbilical cord mesenchymal stem cells, have showed promising results (UC-MSCs). UC-MSCs have been the focus of much study because to their potential as a treatment option for COVID-19 patients. Cytokine release syndrome, often called cytokine storm, increases the risk of morbidity and mortality from COVID-19. It has been established that UC-MSCs may suppress and control both the adaptive and innate immune responses by modulating the release of immunostimulatory cytokines. The purpose of this study is to assess and clarify the use of UC-MSCs for the treatment of ARDS caused by COVID-19.
Collapse
|
5
|
Zhang W, Zhang J, Huang H. Exosomes from adipose-derived stem cells inhibit inflammation and oxidative stress in LPS-acute kidney injury. Exp Cell Res 2022; 420:113332. [PMID: 36084668 DOI: 10.1016/j.yexcr.2022.113332] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 11/04/2022]
Abstract
Acute renal damage presents a significant danger to kidney health. Previous research has found that acute kidney injury shows high levels of oxidative stress and inflammation caused by sepsis. Although mesenchymal stem cells (MSCs) can repair acute kidney injury. However, involvement of MSCs exosomes generated from adipose tissue and bone marrow in lipopolysaccharide-induced acute kidney damage is not clear. LPS (7.5 mg/kg) intraperitoneal injection was used to produce AKI, and 30 min before the LPS administration, adipose-derived MSCs (ADSCs) exosomes (1 × 105 and 5 × 105) and bone marrow-derived MSCs(BMSCs) exosomes (1 × 105 and 5 × 105) were delivered individually. The function of the rat kidney was explored. Inflammation, oxidative stress, and autophagy levels were further investigated. Both adipose-derived and bone marrow-derived MSCs can enhance renal function and structural damage, such as BUN, Creatinine, and cystatin C levels, as well as tubular damage scores. These findings indicate that both adipose-derived MSCs exosomes and bone marrow-derived MSCs exosomes decrease oxidative stress and inflammation, as well as make a substantial influence on kidney tissue in autophagy levels. Furthermore, compared to bone marrow-derived MSCs exosomes, adipose-derived MSCs exosomes improved kidney function and structure more significantly. We discovered that adipose-derived MSCs exosomes protect against LPS-induced AKI by inhibiting oxidative stress and inflammation.
Collapse
Affiliation(s)
- Wen Zhang
- Department of General Practice, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, China
| | - Jian Zhang
- Department of Radiology the First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, China
| | - Hua Huang
- Department of Urology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
6
|
Seo Y, Ahn JS, Shin YY, Oh SJ, Song MH, Kang MJ, Oh JM, Lee D, Kim YH, Lee BC, Shin TH, Kim HS. Mesenchymal stem cells target microglia via galectin-1 production to rescue aged mice from olfactory dysfunction. Biomed Pharmacother 2022; 153:113347. [DOI: 10.1016/j.biopha.2022.113347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/01/2022] Open
|
7
|
Anudeep TC, Jeyaraman M, Muthu S, Rajendran RL, Gangadaran P, Mishra PC, Sharma S, Jha SK, Ahn BC. Advancing Regenerative Cellular Therapies in Non-Scarring Alopecia. Pharmaceutics 2022; 14:pharmaceutics14030612. [PMID: 35335987 PMCID: PMC8953616 DOI: 10.3390/pharmaceutics14030612] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Alopecia or baldness is a common diagnosis in clinical practice. Alopecia can be scarring or non-scarring, diffuse or patchy. The most prevalent type of alopecia is non-scarring alopecia, with the majority of cases being androgenetic alopecia (AGA) or alopecia areata (AA). AGA is traditionally treated with minoxidil and finasteride, while AA is treated with immune modulators; however, both treatments have significant downsides. These drawbacks compel us to explore regenerative therapies that are relatively devoid of adverse effects. A thorough literature review was conducted to explore the existing proven and experimental regenerative treatment modalities in non-scarring alopecia. Multiple treatment options compelled us to classify them into growth factor-rich and stem cell-rich. The growth factor-rich group included platelet-rich plasma, stem cell-conditioned medium, exosomes and placental extract whereas adult stem cells (adipose-derived stem cell-nano fat and stromal vascular fraction; bone marrow stem cell and hair follicle stem cells) and perinatal stem cells (umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), Wharton jelly-derived MSCs (WJ-MSCs), amniotic fluid-derived MSCs (AF-MSCs), and placental MSCs) were grouped into the stem cell-rich group. Because of its regenerative and proliferative capabilities, MSC lies at the heart of regenerative cellular treatment for hair restoration. A literature review revealed that both adult and perinatal MSCs are successful as a mesotherapy for hair regrowth. However, there is a lack of standardization in terms of preparation, dose, and route of administration. To better understand the source and mode of action of regenerative cellular therapies in hair restoration, we have proposed the "À La Mode Classification". In addition, available evidence-based cellular treatments for hair regrowth have been thoroughly described.
Collapse
Affiliation(s)
- Talagavadi Channaiah Anudeep
- Department of Plastic Surgery, Topiwala National Medical College and BYL Nair Ch. Hospital, Mumbai 400008, India;
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- À La Mode Esthétique Studio, Mysuru 570011, India
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Madhan Jeyaraman
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Orthopaedics, Faculty of Medicine—Sri Lalithambigai Medical College and Hospital, Dr MGR Educational and Research Institute, Chennai 600095, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul 624304, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (P.G.); (B.-C.A.)
| | - Prabhu Chandra Mishra
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Shilpa Sharma
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
- Department of Paediatric Surgery, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida 201310, India; (M.J.); (S.M.); (S.K.J.)
- International Association of Stem Cell and Regenerative Medicine (IASRM), New Delhi 110092, India; (P.C.M.); (S.S.)
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, Korea;
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (P.G.); (B.-C.A.)
| |
Collapse
|
8
|
Liu J, Hou Z, Wu J, Liu K, Li D, Gao T, Liu W, An B, Sun Y, Mo F, Wang L, Wang Y, Hao J, Hu B. Infusion of hESC derived Immunity-and-matrix regulatory cells improves cognitive ability in early-stage AD mice. Cell Prolif 2021; 54:e13085. [PMID: 34232542 PMCID: PMC8349653 DOI: 10.1111/cpr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/17/2021] [Accepted: 05/22/2021] [Indexed: 12/20/2022] Open
Abstract
Objectives In this study, we administered immunity‐and‐matrix regulatory cells (IMRCs) via tail vein (IV) and intracerebroventricular (ICV) injection to 3‐month‐old 5×FAD transgenic mice to assess the effects of IMRC transplantation on the behaviour and pathology of early‐stage Alzheimer's disease (AD). Materials and methods Clinical‐grade human embryonic stem cell (hESC)‐derived IMRCs were produced under good manufacturing practice (GMP) conditions. Three‐month‐old 5×FAD mice were administered IMRCs via IV and ICV injection. After 3 months, the mice were subjected to behavioural tests and electrophysiological analysis to evaluate their cognitive function, memory ability and synaptic plasticity. The effect of IMRCs on amyloid‐beta (Aβ)‐related pathology was detected by thioflavin‐S staining and Western blot. Quantitative real‐time PCR, ELISA and immunostaining were used to confirm that IMRCs inhibit neuroinflammation. RNA‐seq analysis was performed to measure changes in gene expression and perform a pathway analysis in response to IMRC treatment. Results IMRC administration via tail vein injection significantly ameliorated cognitive deficits in early‐stage AD (5×FAD) mice. However, no significant change was observed in the characteristic pathology of AD in the ICV group. Plaque analysis revealed that IMRCs did not influence either plaque deposition or BACE1 expression. In addition, IMRCs inhibited inflammatory responses and reduced microglial activation in vivo. Conclusions We have shown that peripheral administration of IMRCs can ameliorate AD pathology and associated cognitive deficits.
Collapse
Affiliation(s)
- Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zongren Hou
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Jun Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Kailun Liu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Da Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Tingting Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Wenjing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Bin An
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Yun Sun
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Liu Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Yukai Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Jie Hao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,National Stem Cell Resource Center, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Jeon HJ, Yoon KA, An ES, Kang TW, Sim YB, Ahn J, Choi EK, Lee S, Seo KW, Kim YB, Kang KS. Therapeutic Effects of Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells Combined with Cartilage Acellular Matrix Mediated Via Bone Morphogenic Protein 6 in a Rabbit Model of Articular Cruciate Ligament Transection. Stem Cell Rev Rep 2021; 16:596-611. [PMID: 32112264 DOI: 10.1007/s12015-020-09958-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Osteoarthritis (OA) is a general joint disease. Cartilage damage is associated with a decrease in the density of chondrocytes. Mesenchymal stem cells (MSCs) differentiate into adipocytes, osteocytes and chondrocytes, and are an excellent source of cell therapy. Cartilage-derived extracellular matrix (ECM) promotes chondrogenesis of MSCs. However, the role of MSCs stimulated by ECM is not well known in OA. The purpose of this study is to determine the role of specific factors generated by the application of ECM and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) in managing OA symptoms. Cartilage acellular matrix (CAM), which is a cartilage-derived ECM, was used to promote the chondrogenesis of UCB-MSCs. Induced MSCs were analyzed using chondrogenic markers (aggrecan, collagen type 2, and SOX9) and bone morphogenic protein 6 (BMP6). BMP6 is known to be involved in early chondrogenesis of MSCs. As a result, treatment with CAM significantly increased the expression of chondrogenic markers and BMP6 in UCB-MSCs. Treatment with recombinant human BMP6 also dramatically increased the levels of chondrogenic markers in UCB-MSCs. In addition, UCB-MSCs and CAM were used to evaluate OA symptom improvement in a rabbit articular cruciate ligament transection (ACLT) model. Application of UCB-MSCs and CAM enhanced not only the structure and synthesis of proteoglycan and collagen type 2 but also anti-inflammatory effects in both rabbit joint and synovial fluid. Moreover, the detection of human cells and involvement of BMP6 were confirmed in rabbit cartilage tissues. This study indicates that therapeutic potential of UCB-MSCs with CAM is mediated via BMP6 in OA.
Collapse
Affiliation(s)
- Hyo-Jin Jeon
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Kyung-Ae Yoon
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Eun Suk An
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Tae-Wook Kang
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Yun-Beom Sim
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Jongchan Ahn
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Ehn-Kyung Choi
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Seunghee Lee
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea
| | - Kwang-Won Seo
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea.
| | - Yun-Bae Kim
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea.
| | - Kyung-Sun Kang
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Seoul, Republic of Korea.
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Köse S, Aerts-Kaya F, Uçkan Çetinkaya D, Korkusuz P. Stem Cell Applications in Lysosomal Storage Disorders: Progress and Ongoing Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1347:135-162. [PMID: 33977438 DOI: 10.1007/5584_2021_639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lysosomal storage disorders (LSDs) are rare inborn errors of metabolism caused by defects in lysosomal function. These diseases are characterized by accumulation of completely or partially degraded substrates in the lysosomes leading to cellular dysfunction of the affected cells. Currently, enzyme replacement therapies (ERTs), treatments directed at substrate reduction (SRT), and hematopoietic stem cell (HSC) transplantation are the only treatment options for LSDs, and the effects of these treatments depend strongly on the type of LSD and the time of initiation of treatment. However, some of the LSDs still lack a durable and curative treatment. Therefore, a variety of novel treatments for LSD patients has been developed in the past few years. However, despite significant progress, the efficacy of some of these treatments remains limited because these therapies are often initiated after irreversible organ damage has occurred.Here, we provide an overview of the known effects of LSDs on stem cell function, as well as a synopsis of available stem cell-based cell and gene therapies that have been/are being developed for the treatment of LSDs. We discuss the advantages and disadvantages of use of hematopoietic stem cell (HSC), mesenchymal stem cell (MSC), and induced pluripotent stem cell (iPSC)-related (gene) therapies. An overview of current research data indicates that when stem cell and/or gene therapy applications are used in combination with existing therapies such as ERT, SRT, and chaperone therapies, promising results can be achieved, showing that these treatments may result in alleviation of existing symptoms and/or prevention of progression of the disease. All together, these studies offer some insight in LSD stem cell biology and provide a hopeful perspective for the use of stem cells. Further development and improvement of these stem cell (gene) combination therapies may greatly improve the current treatment options and outcomes of patients with a LSD.
Collapse
Affiliation(s)
- Sevil Köse
- Department of Medical Biology, Faculty of Medicine, Atilim University, Ankara, Turkey
| | - Fatima Aerts-Kaya
- Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey.,Hacettepe University Center for Stem Cell Research and Development (PEDI-STEM), Ankara, Turkey
| | - Duygu Uçkan Çetinkaya
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Hematology, Hacettepe University Center for Stem Cell Research and Development (PEDI-STEM), Ankara, Turkey.,Department of Stem Cell Sciences, Hacettepe University Graduate School of Health Sciences, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
11
|
Kang I, Yoo JM, Kim D, Kim J, Cho MK, Lee SE, Kim DJ, Lee BC, Lee JY, Kim JJ, Shin N, Choi SW, Lee YH, Ko HS, Shin S, Hong BH, Kang KS. Graphene Quantum Dots Alleviate Impaired Functions in Niemann-Pick Disease Type C in Vivo. NANO LETTERS 2021; 21:2339-2346. [PMID: 33472003 DOI: 10.1021/acs.nanolett.0c03741] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
While the neuropathological characteristics of Niemann-Pick disease type C (NPC) result in a fatal diagnosis, the development of clinically available therapeutic agent remains a challenge. Here we propose graphene quantum dots (GQDs) as a potential candidate for the impaired functions in NPC in vivo. In addition to the previous findings that GQDs exhibit negligible long-term toxicity and are capable of penetrating the blood-brain barrier, GQD treatment reduces the aggregation of cholesterol in the lysosome through expressed physical interactions. GQDs also promote autophagy and restore defective autophagic flux, which, in turn, decreases the atypical accumulation of autophagic vacuoles. More importantly, the injection of GQDs inhibits the loss of Purkinje cells in the cerebellum while also demonstrating reduced activation of microglia. The ability of GQDs to alleviate impaired functions in NPC proves the promise and potential of the use of GQDs toward resolving NPC and other related disorders.
Collapse
Affiliation(s)
- Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Je Min Yoo
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
- BioGraphene Inc., 555 West Fifth Street, Los Angeles, California 90013, United States
| | - Donghoon Kim
- BioGraphene Inc., Advanced Institute of Convergence Technology, Suwon 16229, Korea
| | - Juhee Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Myung Keun Cho
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Seung-Eun Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Dong Jin Kim
- Graphene Square Inc. & Graphene Research Center, Advanced Institute of Convergence Technology, Suwon 16229, Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jin Young Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Nari Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute, Chungcheongbuk-do 28119, Korea
- Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Korea
| | - Han Seok Ko
- Department of Neurology & Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Seokmin Shin
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
| | - Byung Hee Hong
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul 08826, Korea
- BioGraphene Inc., Advanced Institute of Convergence Technology, Suwon 16229, Korea
- Graphene Square Inc. & Graphene Research Center, Advanced Institute of Convergence Technology, Suwon 16229, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
12
|
Kim YJ, Ahn HJ, Lee SH, Lee MH, Kang KS. Effects of conditioned media from human umbilical cord blood-derived mesenchymal stem cells in the skin immune response. Biomed Pharmacother 2020; 131:110789. [PMID: 33152947 DOI: 10.1016/j.biopha.2020.110789] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease in which type 2 allergic inflammation plays a critical role. In this study, the anti-inflammatory effect of conditioned media from human umbilical cord blood-derived mesenchymal stem cells (USC-CM) was investigated in order to apply it as an effective treatment with a low risk of side effects that can overcome the limitations of AD treatment which is currently in use. We found that USC-CM has various growth factors and cytokines associated with anti-inflammatory effect. RT-PCR and ELISA analysis showed that USC-CM inhibited the levels of type 2 cytokine and chemokine Thymus and activation-regulated chemokine (TARC), TNF-α and IL-6 in TNF-α/IFN-γ-stimulated HaCaT cells. In addition, USC-CM inhibited IL-4 and IL-13 levels in Th2 cells. Therefore, the results of our study demonstrated that USC-CM has anti-inflammatory effect in TNF-α/IFN-γ-stimulated HaCaT cells which associated with the inhibition of the immunoglobulin (IgE) secretion by activating B cell line. Our In vivo results showed that when the USC-CM was applied to lesions of patients with the mild AD for 4 weeks, the skin barrier was strengthened by increasing the level of Corneometer and decreasing the value of transepidermal water loss (TEWL). In conclusion, the results suggest that USC-CM may have therapeutic effect for AD as cosmetics and drug materials.
Collapse
Affiliation(s)
- Yoon-Jin Kim
- Derma Science R&D Center, Primoris International CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Hee-Jin Ahn
- Cytotherapy R&D Center, PRIMORIS CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Seung-Hee Lee
- Stem Cells and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., 2nd Floor, Biotechnology Center, #81 Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Mi-Hye Lee
- GMP Center, Kangstem Biotech Co., Ltd., 6nd Floor, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Cytotherapy R&D Center, PRIMORIS CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea.
| |
Collapse
|
13
|
Xu Q, Yan P, Duan XJ, Wu X, Chen XJ, Luo M, Peng JC, Feng LX, Liu J, Zhong HL, Cheng W, Zou QY, Duan SB. Human umbilical cord-derived mesenchymal stem cells and human cord blood mononuclear cells protect against cisplatin-induced acute kidney injury in rat models. Exp Ther Med 2020; 20:145. [PMID: 33093883 PMCID: PMC7571324 DOI: 10.3892/etm.2020.9274] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Human umbilical cord-derived mesenchymal stem cells (hUCMSCs) are a promising tool to attenuate cisplatin (CP)-induced acute kidney injury (AKI). However, whether the transplantation of human cord blood mononuclear cells (hCBMNCs) exhibits similar protective effects and their potential underlying mechanisms of action remain unclear. The present study aimed to determine the protective effects of hUCMSCs and hCBMNCs transplantation therapies on an established CP-induced rat model and explore their underlying mechanisms of action. A total of 24 Sprague-Dawley rats, selected based on body weight, were randomly assigned into 4 groups: i) normal control; ii) model (CP); iii) hCBMNCs (CP + hCBMNCs); and iv) hUCMSCs (CP + hUCMSCs). hUCMSCs (2.0x106 cells) and hCBMNCs (2.0x106 cells) were injected into the femoral vein of rats 24 h after CP (8 mg/kg) treatment. To determine the effects of hCBMNCs and hUCMSCs on CP-induced rats, renal function assessment and histological evaluations were performed. Expression levels of high mobility group box 1 (HMGB1) and the ratio of Bax/Bcl2 in renal tissues were detected to elucidate their underlying molecular mechanisms of action. The results demonstrated that transplantation of hUCMSCs and hCBMNCs significantly improved renal function in CP-induced AKI rats, as evidenced by the enhancement of renal morphology; decreased concentrations of blood urea nitrogen and serum creatinine; and a lower percentage of apoptotic renal tubular cells. The expression of HMGB1 and the ratio of Bax/Bcl-2 were significantly reduced in the hUCMSCs and hCBMNCs groups compared with CP group. In conclusion, the present study indicated that hCBMNCs exert similar protective effects to hUCMSCs on CP-induced AKI. hUCMSCs and hCBMNCs protect against CP-induced AKI by suppressing HMGB1 expression and preventing cell apoptosis.
Collapse
Affiliation(s)
- Qian Xu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Ping Yan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Xiang-Jie Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Xi Wu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Xiao-Jun Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Min Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Jing-Cheng Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Li-Xin Feng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Jie Liu
- Translational Center for Stem Cell Research, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, P.R. China
| | - Hui-Lin Zhong
- Neuromedical Research Center, Guangdong 999 Brain Hospital, Guangzhou, Guangdong 510510, P.R. China
| | - Wei Cheng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| | - Qing-Yan Zou
- Neuromedical Research Center, Guangdong 999 Brain Hospital, Guangzhou, Guangdong 510510, P.R. China
| | - Shao-Bin Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
14
|
Role of Nrf2 in Lipopolysaccharide-Induced Acute Kidney Injury: Protection by Human Umbilical Cord Blood Mononuclear Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6123459. [PMID: 32774680 PMCID: PMC7407026 DOI: 10.1155/2020/6123459] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/24/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
Background Acute kidney injury (AKI) is one of the common complications of sepsis. Heretofore, there is no effective treatment for septic AKI. Recent studies have revealed that besides treating hematological malignancies, human umbilical cord blood mononuclear cells (hUCBMNCs) show good therapeutic effects on other diseases. But whether hUCBMNCs can protect against septic AKI and its underlying mechanism are unknown. Methods The rat model of lipopolysaccharide- (LPS-) induced AKI was developed, and the injection of hUCBMNCs was executed to prevent and treat AKI. ML385, a specific nuclear factor E2-related factor 2 (Nrf2) inhibitor, was used to silence Nrf2. The cell experiments were conducted to elaborate the protective mechanism of Nrf2 pathway. Results An effective model of LPS-induced AKI was established. Compared to the rats only with LPS injection, the levels of inflammation, reactive oxygen species (ROS), and apoptosis in renal tissues after hUCBMNC injection were markedly attenuated. Pathological examination also indicated significant remission of renal tissue injury in the LPS+MNCs group, compared to rats in the LPS group. Transmission electron microscopy (TEM) showed that the damage of the mitochondria in the LPS+MNCs group was lighter than that in the LPS group. Noteworthily, the renal Nrf2/HO-1 pathway was activated and autophagy was enhanced after hUCBMNC injection. ML385 could partly reverse the renoprotective effect of hUCBMNCs, which could demonstrate that Nrf2 participated in the protection of hUCBMNCs. Cell experiments showed that increasing the expression level of Nrf2 could alleviate LPS-induced cell injury by increasing the autophagy level and decreasing the injury of the mitochondria in HK-2 cells. Conclusion All results suggest that hUCBMNCs can protect against LPS-induced AKI via the Nrf2 pathway. Activating Nrf2 can upregulate autophagy to protect LPS-induced cell injury.
Collapse
|
15
|
Kook MG, Lee S, Shin N, Kong D, Kim DH, Kim MS, Kang HK, Choi SW, Kang KS. Repeated intramuscular transplantations of hUCB-MSCs improves motor function and survival in the SOD1 G 93A mice through activation of AMPK. Sci Rep 2020; 10:1572. [PMID: 32005848 PMCID: PMC6994691 DOI: 10.1038/s41598-020-58221-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/06/2020] [Indexed: 11/09/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that is characterized by loss of motor neurons and degeneration of neuromuscular junctions. To improve disease progression, previous studies have suggested many options that have shown beneficial effects in diseases, especially stem cell therapy. In this study, we used repeated intramuscular transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) and observed positive effects on muscle atrophy and oxidative stress. In an in vivo study, motor function, body weight and survival rate were assessed, and skeletal muscle tissues were analyzed by western blotting and immunohistochemistry. After intramuscular transplantation, the hUCB-MSCs survived within the skeletal muscle for at least 1 week. Transplantation ameliorated muscle atrophy and the rate of neuromuscular degeneration in skeletal muscle through reductions in intracellular ROS levels. Both expression of skeletal muscle atrophy markers, muscle atrophy F-box (MAFbx)/atrogin1 and muscle RING finger 1 (MuRF1), were also reduced; however, the reductions were not significant. Moreover, transplantation of hUCB-MSCs improved protein synthesis and inhibited the iNOS/NO signaling pathway through AMPK activation. Our results suggest that repeated intramuscular transplantation of hUCB-MSCs can be a practical option for stem cell therapy for ALS.
Collapse
Affiliation(s)
- Myung Geun Kook
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - SeungEun Lee
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nari Shin
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dasom Kong
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Da-Hyun Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Min-Soo Kim
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyun Kyoung Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Li XW, Feng LX, Zhu XJ, Liu Q, Wang HS, Wu X, Yan P, Duan XJ, Xiao YQ, Cheng W, Peng JC, Zhao F, Deng YH, Duan SB. Human umbilical cord blood mononuclear cells protect against renal tubulointerstitial fibrosis in cisplatin-treated rats. Biomed Pharmacother 2019; 121:109662. [PMID: 31810124 DOI: 10.1016/j.biopha.2019.109662] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 01/18/2023] Open
Abstract
Currently, there is no effective method to prevent renal interstitial fibrosis after acute kidney injury (AKI). In this study, we established and screened a new renal interstitial fibrosis rat model after cisplatin-induced AKI. Our results indicated that rats injected with 4 mg/kg cisplatin once a week for two weeks after firstly administrated with 6.5 mg/kg loading dose of cisplatin could set up a more accurate model reflecting AKI progression to renal interstitial fibrosis. Then, we investigated the effects and possible mechanisms of human umbilical cord blood mononuclear cells (hUCBMNCs) on renal tubular interstitial fibrosis after cisplatin-induced AKI. In rats injected with hUCBMNCs for four times, level of matrix metalloproteinase 7 (MMP-7) in serum and urine, urinary albumin/creatinine ratio, tubular pathological scores, the relative collagen area of the tubulointerstitial region, endoplasmic reticulum dilation and the mitochondrial ultrastructural damage were significantly improved. The level of reactive oxygen species, α-smooth muscle actin (α-SMA), [NOD]-like pyrin domain containing protein 3 and cleaved-Caspase 3 in renal tissue decreased significantly. However, in rats injected with hUCBMNCs for two times, no significant difference was discovered in MMP-7 levels and urinary albumin/creatinine ratio. Although expression of α-SMA and the percentage areas of collagen staining in tubulointerstitial tissues were ameliorated in rats injected with hUCBMNCs for two times, the effects were significantly weaker than those in rats injected with hUCBMNCs for four times. Taken together, our study constructed a highly efficient, duplicable novel rat model of renal fibrosis after cisplatin-induced AKI. Multiple injections of hUCBMNCs may prevent renal interstitial fibrosis after cisplatin-induced AKI.
Collapse
Affiliation(s)
- Xu-Wei Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Li-Xin Feng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Xue-Jing Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Qian Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Hong-Shen Wang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Xi Wu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Ping Yan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Xiang-Jie Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Ye-Qing Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Wei Cheng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Jin-Cheng Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Fei Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Ying-Hao Deng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China
| | - Shao-Bin Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan 410011, China.
| |
Collapse
|
17
|
Li XW, Feng LX, Zhu XJ, Liu Q, Wang HS, Wu X, Yan P, Duan XJ, Xiao YQ, Cheng W, Peng JC, Zhao F, Deng YH, Duan SB. Human umbilical cord blood mononuclear cells protect against renal tubulointerstitial fibrosis in cisplatin-treated rats. Biomed Pharmacother 2019; 121:109310. [PMID: 31710895 DOI: 10.1016/j.biopha.2019.109310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/28/2019] [Accepted: 07/31/2019] [Indexed: 01/18/2023] Open
Abstract
Currently, there is no effective method to prevent renal interstitial fibrosis after acute kidney injury (AKI). In this study, we established and screened a new renal interstitial fibrosis rat model after cisplatin-induced AKI. Our results indicated that rats injected with 4 mg/kg cisplatin once a week for two weeks after firstly administrated with 6.5 mg/kg loading dose of cisplatin could set up a more accurate model reflecting AKI progression to renal interstitial fibrosis. Then, we investigated the effects and possible mechanisms of human umbilical cord blood mononuclear cells (hUCBMNCs) on renal tubular interstitial fibrosis after cisplatin-induced AKI. In rats injected with hUCBMNCs for four times, level of matrix metalloproteinase 7(MMP-7)in serum and urine, urinary albumin/creatinine ratio, tubular pathological scores, the relative collagen area of the tubulointerstitial region, endoplasmic reticulum dilation and the mitochondrial ultrastructural damage were significantly improved. The level of reactive oxygen species, α-smooth muscle actin (α-SMA), [NOD]-like pyrin domain containing protein 3 and cleaved-Caspase 3 in renal tissue decreased significantly. However, in rats injected with hUCBMNCs for two times, no significant difference was discovered in MMP-7 levels and urinary albumin/creatinine ratio. Although expression of α-SMA and the percentage areas of collagen staining in tubulointerstitial tissues were ameliorated in rats injected with hUCBMNCs for two times, the effects were significantly weaker than those in rats injected with hUCBMNCs for four times. Taken together, our study constructed a highly efficient, duplicable novel rat model of renal fibrosis after cisplatin-induced AKI. Multiple injections of hUCBMNCs may prevent renal interstitial fibrosis after cisplatin-induced AKI.
Collapse
Affiliation(s)
- Xu-Wei Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Li-Xin Feng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Xue-Jing Zhu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Qian Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Hong-Shen Wang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Xi Wu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Ping Yan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Xiang-Jie Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Ye-Qing Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Wei Cheng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Jin-Cheng Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Fei Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Ying-Hao Deng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China
| | - Shao-Bin Duan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, 410011, China.
| |
Collapse
|
18
|
Yu Y, Yoo SM, Park HH, Baek SY, Kim YJ, Lee S, Kim YL, Seo KW, Kang KS. Preconditioning with interleukin-1 beta and interferon-gamma enhances the efficacy of human umbilical cord blood-derived mesenchymal stem cells-based therapy via enhancing prostaglandin E2 secretion and indoleamine 2,3-dioxygenase activity in dextran sulfate sodium-induced colitis. J Tissue Eng Regen Med 2019; 13:1792-1804. [PMID: 31293088 DOI: 10.1002/term.2930] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 04/07/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022]
Abstract
Preconditioning with inflammatory cytokines has improved mesenchymal stem cells characteristics, including differentiation and immunomodulating functions. In this study, we developed a preconditioning combination strategy using interleukin-1beta (IL-1β) and interferon-gamma (IFN-γ) to enhance the immuneregulatory ability of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs). Our results showed that hUCB-MSCs preconditioned with IL-1β and IFN-γ (primed hUCB-MSCs) created a statistically significant decrease in peripheral blood mononuclear cell proliferation, indicating that their immunosuppressive ability was increased. The secretion of PGE2, cyclooxygenase 2 mRNA expression, and indoleamine 2,3-dioxygenase (IDO) mRNA expression in primed hUCB-MSCs was significantly higher than those in the untreated hUCB-MSCs or the IL-1β or IFN-γ only treated hUCB-MSCs. When inhibitors of IDO and PGE2 were treated, peripheral blood mononuclear cell proliferation, which is inhibited by primed hUCB-MSCs, was recovered. We found that Th1 T cell differentiation was also inhibited by PGE2 and IDO in the primed hUCB-MSCs, and Tregs differentiation was increased by PGE2 and IDO in the primed hUCB-MSCs. Furthermore, the primed hUCB-MSCs as well as supernatants increase CD4+ T cells migration. We demonstrated the therapeutic effects of primed hUCB-MSCs in dextran sulfate sodium-induced colitis model. In conclusion, we have demonstrated that primed hUCB-MSCs simultaneously enhance PGE2 and IDO and greatly improve the immunoregulatory capacity of MSCs, and we have developed an optimal condition for pretreatment of MSCs for the treatment of immune diseases. Our results raise the possibility that the combination of PGE2 and IDO could be therapeutic mediators for controlling immunosuppression of MSCs.
Collapse
Affiliation(s)
- Yeonsil Yu
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Sae Mi Yoo
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Hwan Hee Park
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Song Yi Baek
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Yoon-Jin Kim
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Yu Lee Kim
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Kwang-Won Seo
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea
| | - Kyung-Sun Kang
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology Center, Seoul National University, Seoul, South Korea.,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
19
|
Yu Y, Yoon K, Kang T, Jeon H, Sim Y, Choe SH, Baek SY, Lee S, Seo K, Kang K. Therapeutic effect of long‐interval repeated intravenous administration of human umbilical cord blood‐derived mesenchymal stem cells in
DBA
/1 mice with collagen‐induced arthritis. J Tissue Eng Regen Med 2019; 13:1134-1142. [DOI: 10.1002/term.2861] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 12/20/2018] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Yeonsil Yu
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Kyung‐Ae Yoon
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Tae‐Wook Kang
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Hyo‐Jin Jeon
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Yun‐Beom Sim
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Seung Hoon Choe
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Song Yi Baek
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Kwang‐Won Seo
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
| | - Kyung‐Sun Kang
- Stem Cell and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., Biotechnology CenterSeoul National University Seoul South Korea
- Adult Stem Cell Research Center, College of Veterinary MedicineSeoul National University Seoul South Korea
- Research Institute for Veterinary Science, College of Veterinary MedicineSeoul National University Seoul South Korea
| |
Collapse
|
20
|
Relaño-Ginés A, Lehmann S, Deville de Périère D, Hirtz C. Dental stem cells as a promising source for cell therapies in neurological diseases. Crit Rev Clin Lab Sci 2019; 56:170-181. [DOI: 10.1080/10408363.2019.1571478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Aroa Relaño-Ginés
- DERBS, Faculty of Odontology, CHU de Montpellier, University of Montpellier, Montpellier, France
| | - Sylvain Lehmann
- LBPC-PPC - IRMB, CHU de Montpellier, University of Montpellier, Montpellier, France
| | - Dominique Deville de Périère
- DERBS, Faculty of Odontology, CHU de Montpellier, University of Montpellier, Montpellier, France
- LBPC-PPC - IRMB, CHU de Montpellier, University of Montpellier, Montpellier, France
| | - Christophe Hirtz
- DERBS, Faculty of Odontology, CHU de Montpellier, University of Montpellier, Montpellier, France
- LBPC-PPC - IRMB, CHU de Montpellier, University of Montpellier, Montpellier, France
| |
Collapse
|
21
|
Stem cell-secreted 14,15- epoxyeicosatrienoic acid rescues cholesterol homeostasis and autophagic flux in Niemann-Pick-type C disease. Exp Mol Med 2018; 50:1-14. [PMID: 30429460 PMCID: PMC6235958 DOI: 10.1038/s12276-018-0176-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/06/2018] [Accepted: 07/18/2018] [Indexed: 12/27/2022] Open
Abstract
We previously demonstrated that the direct transplantation of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) into the dentate gyrus ameliorated the neurological symptoms of Niemann–Pick type C1 (NPC1)-mutant mice. However, the clinical presentation of NPC1-mutant mice was not fully understood with a molecular mechanism. Here, we found 14,15-epoxyeicosatrienoic acid (14,15-EET), a cytochrome P450 (CYP) metabolite, from hUCB-MSCs and the cerebella of NPC1-mutant mice and investigated the functional consequence of this metabolite. Our screening of the CYP2J family indicated a dysregulation in the CYP system in a cerebellar-specific manner. Moreover, in Purkinje cells, CYP2J6 showed an elevated expression level compared to that of astrocytes, granule cells, and microglia. In this regard, we found that one CYP metabolite, 14,15-EET, acts as a key mediator in ameliorating cholesterol accumulation. In confirming this hypothesis, 14,15-EET treatment reduced the accumulation of cholesterol in human NPC1 patient-derived fibroblasts in vitro by suppressing cholesterol synthesis and ameliorating the impaired autophagic flux. We show that the reduced activity within the CYP system in the cerebellum could cause the neurological symptoms of NPC1 patients, as 14,15-EET treatment significantly rescued cholesterol accumulation and impaired autophagy. We also provide evidence that the intranasal administration of hUCB-MSCs is a highly promising alternative to traumatic surgical transplantation for NPC1 patients. An acid secreted by stem cells can reduce the excess cholesterol caused by a genetic metabolic disorder. Niemann–Pick type C disease is a rare, inherited condition that causes defective muscular development and progressive neurological degeneration. A key disease mechanism is the excessive accumulation of cholesterol within cells. Kyung-Sun Kang at Seoul National University, South Korea, and co-workers have demonstrated that a metabolite molecule called 14,15-epoxyeicosatrienoic acid (14,15-EET) derived from stem cells from human umbilical cord blood significantly reduced cholesterol in Neimann-Pick Type C mouse models and human cell samples. The team administered the stem cell therapy non-invasively via the nose, and observed significant improvements in motor function in the mice. Experiments in both animals and cells showed that the treatment resulted in reduced cholesterol levels and the correction of defective signalling within cells.
Collapse
|
22
|
Coronary stents with inducible VEGF/HGF-secreting UCB-MSCs reduced restenosis and increased re-endothelialization in a swine model. Exp Mol Med 2018; 50:1-14. [PMID: 30174328 PMCID: PMC6119684 DOI: 10.1038/s12276-018-0143-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 05/30/2018] [Accepted: 06/07/2018] [Indexed: 12/12/2022] Open
Abstract
Atherosclerotic plaques within the vasculature may eventually lead to heart failure. Currently, cardiac stenting is the most effective and least invasive approach to treat this disease. However, in-stent restenosis is a complex chronic side effect of stenting treatment. This study used coronary stents coated with stem cells secreting angiogenic growth factors via an inducible genome-editing system to reduce stent restenosis and induce re-endothelialization within the artery. The characteristics of the cells and their adhesion properties on the stents were confirmed, and the stents were transplanted into a swine model to evaluate restenosis and the potential therapeutic use of stents with stem cells. Restenosis was evaluated using optical coherence tomography (OCT), microcomputed tomography (mCT) and angiography, and re-endothelialization was evaluated by immunostaining after cardiac stent treatment. Compared to a bare metal stent (BMS) or a parental umbilical cord blood-derived mesenchymal stem cell (UCB-MSC)-coated stent, the stents with stem cells capable of the controlled release of hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) successfully reduced restenosis within the stent and induced natural re-endothelialization. Furthermore, UCB-MSCs exhibited the ability to differentiate into endothelial cells in Matrigel, and HGF and VEGF improved this differentiation. Our study indicates that stents coated with UCB-MSCs secreting VEGF/HGF reduce the restenosis side effects of cardiac stenting with improved re-endothelialization.
Collapse
|
23
|
Bak DH, Choi MJ, Kim SR, Lee BC, Kim JM, Jeon ES, Oh W, Lim ES, Park BC, Kim MJ, Na J, Kim BJ. Human umbilical cord blood mesenchymal stem cells engineered to overexpress growth factors accelerate outcomes in hair growth. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2018; 22:555-566. [PMID: 30181702 PMCID: PMC6115345 DOI: 10.4196/kjpp.2018.22.5.555] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/04/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022]
Abstract
Human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) are used in tissue repair and regeneration; however, the mechanisms involved are not well understood. We investigated the hair growth-promoting effects of hUCB-MSCs treatment to determine whether hUCB-MSCs enhance the promotion of hair growth. Furthermore, we attempted to identify the factors responsible for hair growth. The effects of hUCB-MSCs on hair growth were investigated in vivo, and hUCB-MSCs advanced anagen onset and hair follicle neogeneration. We found that hUCB-MSCs co-culture increased the viability and up-regulated hair induction-related proteins of human dermal papilla cells (hDPCs) in vitro. A growth factor antibody array revealed that secretory factors from hUCB-MSCs are related to hair growth. Insulin-like growth factor binding protein-1 (IGFBP-1) and vascular endothelial growth factor (VEGF) were increased in co-culture medium. Finally, we found that IGFBP-1, through the co-localization of an IGF-1 and IGFBP-1, had positive effects on cell viability; VEGF secretion; expression of alkaline phosphatase (ALP), CD133, and β-catenin; and formation of hDPCs 3D spheroids. Taken together, these data suggest that hUCB-MSCs promote hair growth via a paracrine mechanism.
Collapse
Affiliation(s)
- Dong Ho Bak
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Mi Ji Choi
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Soon Re Kim
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Byung Chul Lee
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Jae Min Kim
- Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| | - Eun Su Jeon
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam 13494, Korea
| | - Wonil Oh
- Biomedical Research Institute, R&D Center, MEDIPOST Co., Ltd., Seongnam 13494, Korea
| | - Ee Seok Lim
- Thema Dermatologic Clinic, Seoul 06524, Korea
| | - Byung Cheol Park
- Department of Dermatology, Dankook Medical College, Cheonan 31116, Korea
| | | | - Jungtae Na
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea
| | - Beom Joon Kim
- Department of Dermatology, College of Medicine, Chung-Ang University, Seoul 06973, Korea.,Department of Medicine, Graduate School, Chung-Ang University, Seoul 06973, Korea
| |
Collapse
|
24
|
Park EH, Lim HS, Lee S, Roh K, Seo KW, Kang KS, Shin K. Intravenous Infusion of Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Rheumatoid Arthritis: A Phase Ia Clinical Trial. Stem Cells Transl Med 2018; 7:636-642. [PMID: 30112846 PMCID: PMC6127229 DOI: 10.1002/sctm.18-0031] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 12/11/2022] Open
Abstract
Based on immunomodulatory actions of human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs), in vitro or preclinical studies of hUCB‐MSCs have been conducted extensively in rheumatoid arthritis (RA). However, few human trials have investigated the outcomes of hUCB‐MSC infusions. The CURE‐iv trial was a phase I, uncontrolled, open label trial for RA patients with moderate disease activity despite treatment with methotrexate. The patients received a single intravenous infusion of 2.5 × 107, 5 × 107, or 1 × 108 cells of hUCB‐MSCs for 30 minutes, three patients in each cluster, with an increment of cell numbers when there was no dose‐limited adverse event. Clinical and safety assessments were performed during the study period, and serum cytokines were measured at baseline and 24 hours after the infusion. Out of 11 screened RA patients, 9 were enrolled. The participants were predominantly female (78%) and the mean age was 57.4 years. The mean disease duration was 9.5 years, and baseline 28‐joint disease activity score (DAS28; using erythrocyte sedimentation rate) was 4.53. There was no major toxicity in all clusters up to 4 weeks after the infusion. Serum erythrocyte sedimentation rate changes at 4 weeks (n = 9) were −7.9 ± 10.4 (p = .0517) and DAS28 changes were −1.60 ± 1.57 (p = .0159). Reduced levels of IL‐1β, IL‐6, IL‐8, and TNF‐α at 24 hours were observed in the cluster infused with 1 × 108 MSCs. This phase Ia hUCB‐MSC infusion trial for established RA patients revealed no short‐term safety concerns. Stem Cells Translational Medicine2018
Collapse
Affiliation(s)
- Eun Hye Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hee-Suk Lim
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Seunghee Lee
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kyounghwan Roh
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kwang-Won Seo
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kyung-Sun Kang
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea.,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kichul Shin
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| |
Collapse
|
25
|
Jung SJ, Kook MG, Kim S, Kang KS, Soh KS. Homing of the Stem Cells from the Acupoint ST-36 to the Site of a Spinal Cord Injury: A Preliminary Study. J Acupunct Meridian Stud 2018; 11:133-136. [PMID: 29879473 DOI: 10.1016/j.jams.2018.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/28/2018] [Accepted: 05/28/2018] [Indexed: 01/16/2023] Open
Abstract
Homing of stem cells (SCs) to desired targets such as injured tissues remains a lingering problem in cell-based therapeutics. Studies on the biodistribution of intravenously administered SCs have shown the inefficacy of blood vessels as the homing path because most of the injected SCs are captured in the capillary beds of the lungs. We considered an alternative administration method using the acupuncture meridians or the primo vascular system. We injected SCs at the acupoint Zusanli (ST-36) below the knee of a nude mouse with a spinal cord injured at the thoracic T9-10 vertebrae. The SCs migrated from the ST-36, along the sciatic nerve, the lumbar 4-5, and then the spinal cord to the injury point T9-10. The SCs were not randomly scattered but were rather well aligned like marathon race runners, along the primo vascular system route toward the injury point. We observed the SCs at 1, 3, 6, 9, 12, and 15 hours after injection. The fast runners among the injected SCs took about 6 hours to reach the sciatic nerve, about 9 hours to reach the lumbar 4-5, and about 15 hours to reach the injury point T9-10.
Collapse
Affiliation(s)
- Sharon Jiyoon Jung
- Graduate School of Convergence Science and Technology, and Advanced Primo Research Laboratory, Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, South Korea; Department of Anatomy, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Myung Geun Kook
- Department of Veterinary Public Health, College of Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Sungchul Kim
- Department of Acupuncture & Moxibustion, Wonkwang University, Gwangju Medical Hospital, Gwangju 61729, South Korea.
| | - Kyung-Sun Kang
- Department of Veterinary Public Health, College of Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| | - Kwang-Sup Soh
- Department of Physics and Astronomy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
26
|
Intrastriatal transplantation of stem cells from human exfoliated deciduous teeth reduces motor defects in Parkinsonian rats. Cytotherapy 2018; 20:670-686. [PMID: 29576501 DOI: 10.1016/j.jcyt.2018.02.371] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 02/11/2018] [Accepted: 02/21/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND This study explored the neural differentiation and therapeutic effects of stem cells from human exfoliated deciduous teeth (SHED) in a rat model of Parkinson's disease (PD). METHODS The SHED were isolated from fresh dental pulp and were induced to differentiate to neurons and dopamine neurons by inhibiting similar mothers against dpp (SMAD) signaling with Noggin and increase conversion of dopamine neurons from SHED with CHIR99021, Sonic Hedgehog (SHH) and FGF8 in vitro. The neural-primed SHED were transplanted to the striatum of 6-hydroxydopamine (6-OHDA)-induced PD rats to evaluate their neural differentiation and functions in vivo. RESULTS These SHED were efficiently differentiated to neurons (62.7%) and dopamine neurons (42.3%) through a newly developed method. After transplantation, the neural-induced SHED significantly improved recovery of the motor deficits of the PD rats. The grafted SHED were differentiated into neurons (61%), including dopamine neurons (22.3%), and integrated into the host rat brain by forming synaptic connections. Patch clamp analysis showed that neurons derived from grafted SHED have the same membrane potential profile as dopamine neurons, indicating these cells are dopamine neuron-like cells. The potential molecular mechanism of SHED transplantation in alleviating motor deficits of the rats is likely to be mediated by neuronal replacement and immune-modulation as we detected the transplanted dopamine neurons and released immune cytokines from SHED. CONCLUSION Using neural-primed SHED to treat PD showed significant restorations of motor deficits in 6-OHDA-induced rats. These observations provide further evidence that SHED can be used for cell-based therapy of PD.
Collapse
|
27
|
Choi SW, Shin JH, Kim JJ, Shin TH, Seo Y, Kim HS, Kang KS. Direct cell fate conversion of human somatic stem cells into cone and rod photoreceptor-like cells by inhibition of microRNA-203. Oncotarget 2018; 7:42139-42149. [PMID: 27283900 PMCID: PMC5173122 DOI: 10.18632/oncotarget.9882] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/12/2016] [Indexed: 12/11/2022] Open
Abstract
Stem cell-based photoreceptor differentiation strategies have been the recent focus of therapies for retinal degenerative diseases. Previous studies utilized embryonic stem (ES) cells and neural retina differentiation cocktails, including DKK1 and Noggin. Here, we show a novel microRNA-mediated strategy of retina differentiation from somatic stem cells, which are potential allogeneic cell sources. Human amniotic epithelial stem cells (AESCs) and umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) treated with a retina differentiation cocktail induced gene expressions of retina development-relevant genes. Furthermore, microRNA-203 (miR-203) is abundantly expressed in human AESCs and human UCB-MSCs. This miR-203 is predicted to target multiple retina development-relevant genes, particularly DKK1, CRX, RORβ, NEUROD1, NRL and THRB. The inhibition of miR-203 induced a retina differentiation of AESCs and UCB-MSCs. Moreover, successive treatments of anti-miR-203 led to the expression of both mature photoreceptor (PR) markers, rhodopsin and opsin. In addition, we determined that CRX, NRL and DKK1 are direct targets of miR-203 using a luciferase assay. Thus, the work presented here suggests that somatic stem cells can potentially differentiate into neural retina cell types when treated with anti-miR-203. They may prove to be a source of both PR subtypes for future allogeneic stem cell-based therapies of non-regenerative retina diseases.
Collapse
Affiliation(s)
- Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae-Hoon Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Pusan National University School of Medicine, Busan 49241, Republic of Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
28
|
Hei WH, Almansoori AA, Sung MA, Ju KW, Seo N, Lee SH, Kim BJ, Kim SM, Jahng JW, He H, Lee JH. Adenovirus vector-mediated ex vivo gene transfer of brain-derived neurotrophic factor (BDNF) tohuman umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) promotescrush-injured rat sciatic nerve regeneration. Neurosci Lett 2017; 643:111-120. [PMID: 28215880 DOI: 10.1016/j.neulet.2017.02.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 02/06/2023]
Abstract
This study was designed toinvestigate the efficacy of adenovirus vector-mediated brain-derived neurotrophic factor (BDNF) ex vivo gene transfer to human umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) in a rat sciatic nerve crush injury model. BDNF protein and mRNA expression after infection was checked through an enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). Male Sprague-Dawley rats (200-250g, 6 weeks old) were distributed into threegroups (n=20 each): the control group, UCB-MSC group, and BDNF-adenovirus infected UCB-MSC (BDNF-Ad+UCB-MSC) group. UCB-MSCs (1×106cells/10μl/rat) or BDNF-Ad+UCB-MSCs (1×106cells/10μl/rat)were transplantedinto the rats at the crush site immediately after sciatic nerve injury. Cell tracking was done with PKH26-labeled UCB-MSCs and BDNF-Ad+UCB-MSCs (1×106cells/10μl/rat). The rats were monitored for 4 weeks post-surgery. Results showed that expression of BDNF at both the protein and mRNA levels was higher inthe BDNF-Ad+UCB-MSC group compared to theUCB-MSC group in vitro.Moreover, BDNF mRNA expression was higher in both UCB-MSC group and BDNF-Ad+ UCB-MSC group compared tothe control group, and BDNF mRNA expression in theBDNF-Ad+UCB-MSC group was higher than inboth other groups 5days after surgeryin vivo. Labeled neurons in the dorsal root ganglia (DRG), axon counts, axon density, and sciatic function index were significantly increased in the UCB-MSC and BDNF-Ad+ UCB-MSCgroupscompared to the controlgroup four weeksaftercell transplantation. Importantly,the BDNF-Ad+UCB-MSCgroup exhibited more peripheral nerve regeneration than the other two groups.Our results indicate thatboth UCB-MSCs and BDNF-Ad+UCB-MSCscan improve rat sciatic nerve regeneration, with BDNF-Ad+UCB-MSCsshowing a greater effectthan UCB-MSCs.
Collapse
Affiliation(s)
- Wei-Hong Hei
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea; The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Akram A Almansoori
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea; Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Mi-Ae Sung
- Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Won Ju
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea; Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Nari Seo
- Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sung-Ho Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea; Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Bong-Ju Kim
- Clinical Translational Research Center for Dental Science (CTRC), Seoul National University Dental Hospital, Seoul, Republic of Korea
| | - Soung-Min Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jeong Won Jahng
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Hong He
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Orthodontics, School and hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Jong-Ho Lee
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul, Republic of Korea; Dental Research Institute, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Cho HM, Kim PH, Chang HK, Shen YM, Bonsra K, Kang BJ, Yum SY, Kim JH, Lee SY, Choi MC, Kim HH, Jang G, Cho JY. Targeted Genome Engineering to Control VEGF Expression in Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells: Potential Implications for the Treatment of Myocardial Infarction. Stem Cells Transl Med 2017; 6:1040-1051. [PMID: 28186692 PMCID: PMC5442764 DOI: 10.1002/sctm.16-0114] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/11/2016] [Accepted: 09/01/2016] [Indexed: 12/19/2022] Open
Abstract
Human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs) exhibit potency for the regeneration of infarcted hearts. Vascular endothelial growth factor (VEGF) is capable of inducing angiogenesis and can boost stem cell‐based therapeutic effects. However, high levels of VEGF can cause abnormal blood vessel growth and hemangiomas. Thus, a controllable system to induce therapeutic levels of VEGF is required for cell therapy. We generated an inducible VEGF‐secreting stem cell (VEGF/hUCB‐MSC) that controls the expression of VEGF and tested the therapeutic efficacy in rat myocardial infarction (MI) model to apply functional stem cells to MI. To introduce the inducible VEGF gene cassette into a safe harbor site of the hUCB‐MSC chromosome, the transcription activator‐like effector nucleases system was used. After confirming the integration of the cassette into the locus, VEGF secretion in physiological concentration from VEGF/hUCB‐MSCs after doxycycline (Dox) induction was proved in conditioned media. VEGF secretion was detected in mice implanted with VEGF/hUCB‐MSCs grown via a cell sheet system. Vessel formation was induced in mice transplanted with Matrigel containing VEGF/hUCB‐MSCs treated with Dox. Moreover, seeding of the VEGF/hUCB‐MSCs onto the cardiac patch significantly improved the left ventricle ejection fraction and fractional shortening in a rat MI model upon VEGF induction. Induced VEGF/hUCB‐MSC patches significantly decreased the MI size and fibrosis and increased muscle thickness, suggesting improved survival of cardiomyocytes and protection from MI damage. These results suggest that our inducible VEGF‐secreting stem cell system is an effective therapeutic approach for the treatment of MI. Stem Cells Translational Medicine2017;6:1040–1051
Collapse
Affiliation(s)
- Hyun-Min Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Pyung-Hwan Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyun-Kyung Chang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Yi-Ming Shen
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kwaku Bonsra
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Byung-Jae Kang
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Soo-Young Yum
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Joo-Hyun Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - So-Yeong Lee
- Department of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Min-Cheol Choi
- Department of Veterinary Radiology, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Hyongbum Henry Kim
- Department of Pharmacology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Goo Jang
- Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
30
|
Targeting antioxidant enzyme expression as a therapeutic strategy for ischemic stroke. Neurochem Int 2016; 107:23-32. [PMID: 28043837 DOI: 10.1016/j.neuint.2016.12.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/05/2016] [Accepted: 12/17/2016] [Indexed: 11/24/2022]
Abstract
During ischemic stroke, neurons and glia are subjected to damage during the acute and neuroinflammatory phases of injury. Production of reactive oxygen species (ROS) from calcium dysregulation in neural cells and the invasion of activated immune cells are responsible for stroke-induced neurodegeneration. Scientists have failed thus far to identify antioxidant-based drugs that can enhance neural cell survival and improve recovery after stroke. However, several groups have demonstrated success in protecting against stroke by increasing expression of antioxidant enzymes in neural cells. These enzymes, which include but are not limited to enzymes in the glutathione peroxidase, catalase, and superoxide dismutase families, degrade ROS that otherwise damage cellular components such as DNA, proteins, and lipids. Several groups have identified cellular therapies including neural stem cells and human umbilical cord blood cells, which exert neuroprotective and oligoprotective effects through the release of pro-survival factors that activate PI3K/Akt signaling to upregulation of antioxidant enzymes. Other studies demonstrate that treatment with soluble factors released by these cells yield similar changes in enzyme expression after stroke. Treatment with the cytokine leukemia inhibitory factor increases the expression of peroxiredoxin IV and metallothionein III in glia and boosts expression of superoxide dismutase 3 in neurons. Through cell-specific upregulation of these enzymes, LIF and other Akt-inducing factors have the potential to protect multiple cell types against damage from ROS during the early and late phases of ischemic damage.
Collapse
|
31
|
Shin TH, Kim HS, Kang TW, Lee BC, Lee HY, Kim YJ, Shin JH, Seo Y, Won Choi S, Lee S, Shin K, Seo KW, Kang KS. Human umbilical cord blood-stem cells direct macrophage polarization and block inflammasome activation to alleviate rheumatoid arthritis. Cell Death Dis 2016; 7:e2524. [PMID: 28005072 PMCID: PMC5260999 DOI: 10.1038/cddis.2016.442] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/04/2016] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis (RA) is a long-lasting intractable autoimmune disorder, which has become a substantial public health problem. Despite widespread use of biologic drugs, there have been uncertainties in efficacy and long-term safety. Mesenchymal stem cells (MSCs) have been suggested as a promising alternative for the treatment of RA because of their immunomodulatory properties. However, the precise mechanisms of MSCs on RA-related immune cells are not fully elucidated. The aim of this study was to investigate the therapeutic potential of human umbilical cord blood-derived MSCs (hUCB-MSCs) as a new therapeutic strategy for patients with RA and to explore the mechanisms underlying hUCB-MSC-mediated immunomodulation. Mice with collagen-induced arthritis (CIA) were administered with hUCB-MSCs after the onset of disease, and therapeutic efficacy was assessed. Systemic delivery of hUCB-MSCs significantly ameliorated the severity of CIA to a similar extent observed in the etanercept-treated group. hUCB-MSCs exerted this therapeutic effect by regulating macrophage function. To verify the regulatory effects of hUCB-MSCs on macrophages, macrophages were co-cultured with hUCB-MSCs. The tumor necrosis factor (TNF)-α-mediated activation of cyclooxygenase-2 and TNF-stimulated gene/protein 6 in hUCB-MSCs polarized naive macrophages toward an M2 phenotype. In addition, hUCB-MSCs down-regulated the activation of nucleotide-binding domain and leucine-rich repeat pyrin 3 inflammasome via a paracrine loop of interleukin-1β signaling. These immune-balancing effects of hUCB-MSCs were reproducible in co-culture experiments using peripheral blood mononuclear cells from patients with active RA. hUCB-MSCs can simultaneously regulate multiple cytokine pathways in response to pro-inflammatory cytokines elevated in RA microenvironment, suggesting that treatment with hUCB-MSCs could be an attractive candidate for patients with treatment-refractory RA.
Collapse
Affiliation(s)
- Tae-Hoon Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Tae-Wook Kang
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biomedical Science Building, Seoul National University, Seoul, Republic of Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwa-Yong Lee
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biomedical Science Building, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Jin Kim
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biomedical Science Building, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seunghee Lee
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biomedical Science Building, Seoul National University, Seoul, Republic of Korea
| | - Kichul Shin
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Kwang-Won Seo
- Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biomedical Science Building, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
32
|
Lee YS, Sah SK, Lee JH, Seo KW, Kang KS, Kim TY. Human umbilical cord blood-derived mesenchymal stem cells ameliorate psoriasis-like skin inflammation in mice. Biochem Biophys Rep 2016; 9:281-288. [PMID: 28956015 PMCID: PMC5614481 DOI: 10.1016/j.bbrep.2016.10.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/16/2016] [Accepted: 10/04/2016] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) inhibit the proliferation or activation of lymphocytes, and their inhibitory effects do not require human leukocyte antigen (HLA)-matching because MSCs express low levels of HLA molecules. Therefore, MSCs may be able to regulate immune responses. In this study, we determined whether MSCs could inhibit psoriasis-like skin inflammation in mice. After induction of psoriasis-like skin inflammation using intradermal injection of IL-23 or topical application of imiquimod with or without treatment with MSC, mouse skins were collected, and H&E staining and real-time PCR were performed. IL-23-induced skin inflammation was inhibited when MSCs were injected on day −1 and day 7. The expression of proinflammatory cytokines such as IL-6, IL-17, and TNF-α was inhibited by MSC injection, and the expression of chemokines such as CCL17, CCL20, and CCL27 was also decreased in mouse skin. We also determined whether MSCs could not only prevent but also treat psoriasis-like skin inflammation in mice. Furthermore, in vitro experiments also showed anti-inflammatory effects of MSCs. Dendritic cells which are co-cultured with MSCs suppressed CD4+ T cell activation and differentiation, which are important for the pathogenesis of psoriasis. These results suggest that MSCs could be useful for treating psoriasis. Mesenchymal stem cells inhibit psoriasis-like skin inflammation in mice. Mesenchymal stem cells modulate dendritic cell function. Dendritic cells that co-cultured with mesenchymal stem cells regulate CD4+ T cell differentiation.
Collapse
Affiliation(s)
- Yun Sang Lee
- Department of Dermatology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-040, South Korea
| | - Shyam Kishor Sah
- Department of Dermatology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-040, South Korea
| | - Ji Hyun Lee
- Department of Dermatology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-040, South Korea
| | - Kwang-Won Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea.,Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, Biotechnology Incubating center, Seoul National University, Seoul 151-742, South Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea.,Researh Institute for Veterinary Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Tae-Yoon Kim
- Department of Dermatology, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-040, South Korea
| |
Collapse
|
33
|
Seo Y, Kim HS, Kang I, Choi SW, Shin TH, Shin JH, Lee BC, Lee JY, Kim JJ, Kook MG, Kang KS. Cathepsin S contributes to microglia-mediated olfactory dysfunction through the regulation of Cx3cl1-Cx3cr1 axis in a Niemann-Pick disease type C1 model. Glia 2016; 64:2291-2305. [PMID: 27687148 DOI: 10.1002/glia.23077] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 09/12/2016] [Accepted: 09/15/2016] [Indexed: 12/28/2022]
Abstract
Microglia can aggravate olfactory dysfunction by mediating neuronal death in the olfactory bulb (OB) of a murine model of Niemann-Pick disease type C1 (NPC1), a fatal neurodegenerative disorder accompanied by lipid trafficking defects. In this study, we focused on the crosstalk between neurons and microglia to elucidate the mechanisms underlying extensive microgliosis in the NPC1-affected brain. Microglia in the OB of NPC1 mice strongly expressed CX3C chemokine receptor 1 (Cx3cr1), a specific receptor for the neural chemokine C-X3-C motif ligand 1 (Cx3cl1). In addition, a high level of Cx3cl1 was detected in NPC1 mouse-derived CSF due to enhanced catalytic activity of Cathepsin S (Ctss), which is responsible for Cx3cl1 secretion. Notably, nasal delivery of Cx3cl1 neutralizing antibody or Ctss inhibitor could inhibit the Cx3cl1-Cx3cr1 interaction and support neuronal survival through the suppression of microglial activation, leading to an improvement in the olfactory function in NPC1 mice. Relevant in vitro experiments revealed that intracellular cholesterol accumulation could act as a strong inducer of abnormal Ctss activation and, in turn, stimulated the Cx3cl1-Cx3cr1 axis in microglia via p38 mitogen-activated protein kinase signaling. Our data address the significance of Cx3cl1-Cx3cr1 interaction in the development of microglial neurotoxicity and suggest that Ctss is a key upstream regulator. Therefore, this study contributes to a better understanding of the crosstalk between neurons and microglia in the development of the neurodegeneration and provides a new perspective for the management of olfactory deficits and other microglia-dependent neuropathies. GLIA 2016;64:2291-2305.
Collapse
Affiliation(s)
- Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,School of Medicine, Pusan National University, Busan, 49241, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, South Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,School of Medicine, Pusan National University, Busan, 49241, South Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, South Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Tae-Hoon Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Ji-Hee Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Byung-Chul Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jin Young Lee
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Jae-Jun Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Myung Geun Kook
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea. .,Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
34
|
Kim HS, Yun JW, Shin TH, Lee SH, Lee BC, Yu KR, Seo Y, Lee S, Kang TW, Choi SW, Seo KW, Kang KS. Human umbilical cord blood mesenchymal stem cell-derived PGE2 and TGF-β1 alleviate atopic dermatitis by reducing mast cell degranulation. Stem Cells 2016; 33:1254-66. [PMID: 25522163 DOI: 10.1002/stem.1913] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 10/29/2014] [Accepted: 10/31/2014] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cell (MSC) is a promising tool for the therapy of immune disorders. However, their efficacy and mechanisms in treating allergic skin disorders are less verified. We sought to investigate the therapeutic efficacy of human umbilical cord blood-derived MSCs (hUCB-MSCs) against murine atopic dermatitis (AD) and to explore distinct mechanisms that regulate their efficacy. AD was induced in mice by the topical application of Dermatophagoides farinae. Naïve or activated-hUCB-MSCs were administered to mice, and clinical severity was determined. The subcutaneous administration of nucleotide-binding oligomerization domain 2 (NOD2)-activated hUCB-MSCs exhibited prominent protective effects against AD, and suppressed the infiltration and degranulation of mast cells (MCs). A β-hexosaminidase assay was performed to evaluate the effect of hUCB-MSCs on MC degranulation. NOD2-activated MSCs reduced the MC degranulation via NOD2-cyclooxygenase-2 signaling. In contrast to bone marrow-derived MSCs, hUCB-MSCs exerted a cell-to-cell contact-independent suppressive effect on MC degranulation through the higher production of prostaglandin E2 (PGE2 ). Additionally, transforming growth factor (TGF)-β1 production from hUCB-MSCs in response to interleukin-4 contributed to the attenuation of MC degranulation by downregulating FcεRI expression in MCs. In conclusion, the subcutaneous application of NOD2-activated hUCB-MSCs can efficiently ameliorate AD, and MSC-derived PGE2 and TGF-β1 are required for the inhibition of MC degranulation.
Collapse
Affiliation(s)
- Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine#406 Biotechnology Incubating Center, Seoul National University, Seoul, South Korea; Institute for Stem Cell and Regenerative Medicine in Kangstem Biotech, #406 Biotechnology Incubating Center, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Riches Z, Walia G, Berman JM, Wright TE, Collier AC. ATP-binding cassette proteins BCRP, MRP1 and P-gp expression and localization in the human umbilical cord. Xenobiotica 2015; 46:548-56. [PMID: 26407213 DOI: 10.3109/00498254.2015.1091118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. The umbilical cord is a direct conduit to the fetus hence transporters could have roles in partitioning substances between the maternal-placental-fetal units. Here we determined the expression and localization of the ATP-Binding Cassette (ABC) transporters BCRP (ABCG2), P-gp (ABCB1) and MRP1 (ABCC1) in human umbilical cords. 2. The mRNA for BCRP and MRP1 was detected in 25/25 samples, but P-gp was detected in only 5/25. ABC transporter mRNA expression relative to 18S was 25.6 ± 0.3, 26.5 ± 0.6 and 22.2 ± 0.2 cycles for BCRP, MRP1 and P-gp respectively. 3. Using a subset of 10 umbilical cords, BCRP protein was present in all samples (immunoblot) with positive correlation between mRNA and proteins (p = 0.07, r = 0.62) and between immunoblotting and immunohistochemistry (IHC) (p = 0.03, r = 0.67). P-gp protein was observed in 4/10 samples by both immunoblot and IHC, with no correlation between mRNA and protein (p = 0.45, r = 0.55) or immunoblotting and IHC (p = 0.2, r = 0.72), likely due to small sample size. MRP1 protein was not observed. 4. Localization of BCRP and P-gp proteins was to Wharton's jelly with no specific staining in arterial or venous endothelia. 5. Understanding ABC transporter expression in the umbilical cord may be useful for determining fetal exposures to xenobiotics if functional properties can be defined.
Collapse
Affiliation(s)
- Zoe Riches
- a Faculty of Pharmaceutical Sciences , University of British Columbia , Vancouver , BC , Canada and
| | - Gurinder Walia
- a Faculty of Pharmaceutical Sciences , University of British Columbia , Vancouver , BC , Canada and
| | - Jacob M Berman
- a Faculty of Pharmaceutical Sciences , University of British Columbia , Vancouver , BC , Canada and
| | - Tricia E Wright
- b Department of Obstetrics , Gynecology and Women's Health, John A. Burns School of Medicine, Kapi'Olani Medical Center for Women and Children , Honolulu , HI , USA
| | - Abby C Collier
- a Faculty of Pharmaceutical Sciences , University of British Columbia , Vancouver , BC , Canada and
| |
Collapse
|
36
|
Park GY, Lee EM, Seo MS, Seo YJ, Oh JS, Son WC, Kim KS, Kim JS, Kang JK, Kang KS. Preserved Hippocampal Glucose Metabolism on 18F-FDG PET after Transplantation of Human Umbilical Cord Blood-derived Mesenchymal Stem Cells in Chronic Epileptic Rats. J Korean Med Sci 2015; 30:1232-40. [PMID: 26339161 PMCID: PMC4553668 DOI: 10.3346/jkms.2015.30.9.1232] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 05/18/2015] [Indexed: 11/20/2022] Open
Abstract
Human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) may be a promising modality for treating medial temporal lobe epilepsy. (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) is a noninvasive method for monitoring in vivo glucose metabolism. We evaluated the efficacy of hUCB-MSCs transplantation in chronic epileptic rats using FDG-PET. Rats with recurrent seizures were randomly assigned into three groups: the stem cell treatment (SCT) group received hUCB-MSCs transplantation into the right hippocampus, the sham control (ShC) group received same procedure with saline, and the positive control (PC) group consisted of treatment-negative epileptic rats. Normal rats received hUCB-MSCs transplantation acted as the negative control (NC). FDG-PET was performed at pre-treatment baseline and 1- and 8-week posttreatment. Hippocampal volume was evaluated and histological examination was done. In the SCT group, bilateral hippocampi at 8-week after transplantation showed significantly higher glucose metabolism (0.990 ± 0.032) than the ShC (0.873 ± 0.087; P < 0.001) and PC groups (0.858 ± 0.093; P < 0.001). Histological examination resulted that the transplanted hUCB-MSCs survived in the ipsilateral hippocampus and migrated to the contralateral hippocampus but did not differentiate. In spite of successful engraftment, seizure frequency among the groups was not significantly different. Transplanted hUCB-MSCs can engraft and migrate, thereby partially restoring bilateral hippocampal glucose metabolism. The results suggest encouraging effect of hUCB-MSCs on restoring epileptic networks.
Collapse
Affiliation(s)
- Ga Young Park
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- The Asan Institute for Life Science, Seoul, Korea
| | - Eun Mi Lee
- Department of Neurology, Ulsan University Hospital, Ulsan, Korea
| | - Min-Soo Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Yoo-Jin Seo
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jungsu S. Oh
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Woo-Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ki Soo Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Joong Koo Kang
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
37
|
Therapeutic potential of human umbilical cord blood mesenchymal stem cells on erectile function in rats with cavernous nerve injury. Biotechnol Lett 2015; 37:1515-25. [DOI: 10.1007/s10529-015-1816-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/12/2015] [Indexed: 10/23/2022]
|
38
|
Seo Y, Kim HS, Shin Y, Kang I, Choi SW, Yu KR, Seo KW, Kang KS. Excessive microglial activation aggravates olfactory dysfunction by impeding the survival of newborn neurons in the olfactory bulb of Niemann-Pick disease type C1 mice. Biochim Biophys Acta Mol Basis Dis 2014; 1842:2193-203. [PMID: 25132229 DOI: 10.1016/j.bbadis.2014.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 08/07/2014] [Accepted: 08/08/2014] [Indexed: 12/26/2022]
Abstract
Progressive olfactory impairment is one of the earliest markers of neurodegeneration. However, the underlying mechanism for this dysfunction remains unclear. The present study investigated the possible role of microgliosis in olfactory deficits using a mouse model of Niemann-Pick disease type C1 (NPC1), which is an incurable neurodegenerative disorder with disrupted lipid trafficking. At 7weeks of age, NPC1 mutants showed a distinct olfactory impairment in an olfactory test compared with age-matched wild-type controls (WT). The marked loss of olfactory sensory neurons within the NPC1 affected olfactory bulb (NPC1-OB) suggests that NPC1 dysfunction impairs olfactory structure. Furthermore, the pool of neuroblasts in the OB was diminished in NPC1 mice despite the intact proliferative capacity of neural stem/progenitor cells in the subventricular zone. Instead, pro-inflammatory proliferating microglia accumulated extensively in the NPC1-OB as the disease progressed. To evaluate the impact of abnormal microglial activation on olfaction in NPC1 mice, a microglial inhibition study was performed using the anti-inflammatory agent Cyclosporin A (CsA). Importantly, long-term CsA treatment in NPC1 mice reduced reactive microgliosis, restored the survival of newly generated neurons in the OB and improved overall performance on the olfactory test. Therefore, our study highlights the possible role of microglia in the regulation of neuronal turnover in the OB and provides insight into the possible therapeutic applications of microglial inhibition in the attenuation or reversal of olfactory impairment.
Collapse
Affiliation(s)
- Yoojin Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Institute for Stem Cell and Regenerative Medicine at Kangstem Biotech, Biotechnology Incubating Center, Seoul National University, Seoul 151-742, South Korea
| | - Yooyoung Shin
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Insung Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Kyung-Rok Yu
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea
| | - Kwang-Won Seo
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Institute for Stem Cell and Regenerative Medicine at Kangstem Biotech, Biotechnology Incubating Center, Seoul National University, Seoul 151-742, South Korea.
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 151-742, South Korea.
| |
Collapse
|
39
|
Kang BJ, Kim H, Lee SK, Kim J, Shen Y, Jung S, Kang KS, Im SG, Lee SY, Choi M, Hwang NS, Cho JY. Umbilical-cord-blood-derived mesenchymal stem cells seeded onto fibronectin-immobilized polycaprolactone nanofiber improve cardiac function. Acta Biomater 2014; 10:3007-17. [PMID: 24657671 DOI: 10.1016/j.actbio.2014.03.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 03/08/2014] [Accepted: 03/10/2014] [Indexed: 12/28/2022]
Abstract
Stem cells seeded onto biofunctional materials have greater potency for therapeutic applications. We investigated whether umbilical-cord-blood-derived mesenchymal stem cell (UCB-MSC)-seeded fibronectin (FN)-immobilized polycaprolactone (PCL) nanofibers could improve cardiac function and inhibit left ventricle (LV) remodeling in a rat model of myocardial infarction (MI). Aligned nanofibers were uniformly coated with poly(glycidyl methacrylate) by initiated chemical vapor deposition followed by covalent immobilization of FN proteins. The degree of cell elongation and adhesion efficacy were improved by FN immobilization. Furthermore, genes related to angiogenesis and mesenchymal differentiations were up-regulated in the FN-immobilized PCL nanofibers in comparison to control PCL nanofibers in vitro. 4 weeks after the transplantation in the rat MI model, the echocardiogram showed that the UCB-MSC-seeded FN-immobilized PCL nanofiber group increased LV ejection fraction and fraction shortening as compared to the non-treated control and acellular FN-immobilized PCL nanofiber groups. Histological analysis indicated that the implantation of UCB-MSCs with FN-immobilized PCL nanofibers induced a decrease in MI size and fibrosis, and an increase in scar thickness. This study indicates that FN-immobilized biofunctional PCL nanofibers could be an effective carrier for UCB-MSC transplantation for the treatment of MI.
Collapse
Affiliation(s)
- Byung-Jae Kang
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Hwan Kim
- School of Chemical and Biological Engineering, BioMAX Institute, Seoul National University, Seoul, Republic of Korea
| | - Seul Ki Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Joohyun Kim
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yiming Shen
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sunyoung Jung
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Technology, Daejeon, Republic of Korea
| | - So Yeong Lee
- Laboratory of Veterinary Pharmacology, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Mincheol Choi
- Department of Veterinary Medical Imaging, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, BioMAX Institute, Seoul National University, Seoul, Republic of Korea.
| | - Je-Yoel Cho
- Department of Veterinary Biochemistry, BK21 Plus and Research Institute of Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
40
|
Phinney DG, Isakova IA. Mesenchymal stem cells as cellular vectors for pediatric neurological disorders. Brain Res 2014; 1573:92-107. [PMID: 24858930 DOI: 10.1016/j.brainres.2014.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/07/2014] [Accepted: 05/16/2014] [Indexed: 12/15/2022]
Abstract
Lysosomal storage diseases are a heterogeneous group of hereditary disorders characterized by a deficiency in lysosomal function. Although these disorders differ in their etiology and phenotype those that affect the nervous system generally manifest as a profound deterioration in neurologic function with age. Over the past several decades implementation of various treatment regimens including bone marrow and cord blood cell transplantation, enzyme replacement, and substrate reduction therapy have proved effective for managing some clinical manifestations of these diseases but their ability to ameliorate neurologic complications remains unclear. Consequently, there exists a need to develop alternative therapies that more effectively target the central nervous system. Recently, direct intracranial transplantation of tissue-specific stem and progenitor cells has been explored as a means to reconstitute metabolic deficiencies in the CNS. In this chapter we discuss the merits of bone marrow-derived mesenchymal stem cells (MSCs) for this purpose. Originally identified as progenitors of connective tissue cell lineages, recent findings have revealed several novel aspects of MSC biology that make them attractive as therapeutic agents in the CNS. We relate these advances in MSC biology to their utility as cellular vectors for treating neurologic sequelae associated with pediatric neurologic disorders.
Collapse
Affiliation(s)
- Donald G Phinney
- Department of Molecular Therapeutics, The Scripps Research Institute, 130 Scripps Way, A213, Jupiter, FL 33458, USA.
| | - Iryna A Isakova
- Division of Clinical Laboratory Diagnostics, Biology Department, National Dnepropetrovsk University, Dnepropetrovsk, Ukraine
| |
Collapse
|
41
|
Donepezil Enhances Purkinje Cell Survival and Alleviates Motor Dysfunction by Inhibiting Cholesterol Synthesis in a Murine Model of Niemann Pick Disease Type C. J Neuropathol Exp Neurol 2014; 73:234-43. [DOI: 10.1097/nen.0000000000000045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
42
|
Kim HS, Shin TH, Lee BC, Yu KR, Seo Y, Lee S, Seo MS, Hong IS, Choi SW, Seo KW, Núñez G, Park JH, Kang KS. Human umbilical cord blood mesenchymal stem cells reduce colitis in mice by activating NOD2 signaling to COX2. Gastroenterology 2013; 145:1392-403.e1-8. [PMID: 23973922 DOI: 10.1053/j.gastro.2013.08.033] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 08/14/2013] [Accepted: 08/15/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Decreased levels or function of nucleotide-binding oligomerization domain 2 (NOD2) are associated with Crohn's disease. NOD2 regulates intestinal inflammation, and also is expressed by human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs), to regulate their differentiation. We investigated whether NOD2 is required for the anti-inflammatory activities of MSCs in mice with colitis. METHODS Colitis was induced in mice by administration of dextran sulfate sodium or trinitrobenzene sulfonic acid. Mice then were given intraperitoneal injections of NOD2-activated hUCB-MSCs; colon tissues and mesenteric lymph nodes were collected for histologic analyses. A bromodeoxyuridine assay was used to determine the ability of hUCB-MSCs to inhibit proliferation of human mononuclear cells in culture. RESULTS Administration of hUCB-MSCs reduced the severity of colitis in mice. The anti-inflammatory effects of hUCB-MSCs were greatly increased by activation of NOD2 by its ligand, muramyl dipeptide (MDP). Administration of NOD2-activated hUCB-MSCs increased anti-inflammatory responses in colons of mice, such as production of interleukin (IL)-10 and infiltration by T regulatory cells, and reduced production of inflammatory cytokines. Proliferation of mononuclear cells was inhibited significantly by co-culture with hUCB-MSCs that had been stimulated with MDP. MDP induced prolonged production of prostaglandin (PG)E2 in hUCB-MSCs via the NOD2-RIP2 pathway, which suppressed proliferation of mononuclear cells derived from hUCB. PGE2 produced by hUCB-MSCs in response to MDP increased production of IL-10 and T regulatory cells. In mice, production of PGE2 by MSCs and subsequent production of IL-10 were required to reduce the severity of colitis. CONCLUSIONS Activation of NOD2 is required for the ability of hUCB-MSCs to reduce the severity of colitis in mice. NOD2 signaling increases the ability of these cells to suppress mononuclear cell proliferation by inducing production of PGE2.
Collapse
Affiliation(s)
- Hyung-Sik Kim
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hong IS, Lee HY, Choi SW, Kim HS, Yu KR, Seo Y, Jung JW, Kang KS. The effects of hedgehog on RNA binding protein Msi1 during the osteogenic differentiation of human cord blood-derived mesenchymal stem cells. Bone 2013; 56:416-25. [PMID: 23880227 DOI: 10.1016/j.bone.2013.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 07/09/2013] [Accepted: 07/15/2013] [Indexed: 10/26/2022]
Abstract
Human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) are useful tools for regenerative medicine due to their capacity for self-renewal and multi-lineage differentiation. The appropriate clinical application of MSCs for regenerative medicine requires an integrated understanding of multiple signaling pathways that regulate cell proliferation, stemness and differentiation. However, the potential molecular mechanisms mediating these functions are not completely understood. The effects of hedgehog (Hh) signaling on the osteogenic differentiation of MSCs are still controversial, and the underlying mechanisms are unclear. In the present study, we evaluated the direct effects of Hh signaling on the osteogenic differentiation of hUCB-MSCs and investigated potential downstream regulatory mechanisms responsible for Hh signaling. We observed that Hh signaling acts as a negative regulator of osteogenic differentiation through the suppression of RNA-binding Msi1, which in turn suppresses the expression of Wnt1 and the miR-148 family, especially miR-148b. Moreover, Hh and Msi1 are considered to be potential stemness markers of hUCB-MSCs due to their differentiation-dependent expression profiles. This study provides new insights into mechanisms regulating MSC differentiation and may have implications for a variety of therapeutic applications in the clinic.
Collapse
Affiliation(s)
- In-Sun Hong
- Adult Stem cell Research Center, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Hovakimyan M, Maass F, Petersen J, Holzmann C, Witt M, Lukas J, Frech MJ, Hübner R, Rolfs A, Wree A. Combined therapy with cyclodextrin/allopregnanolone and miglustat improves motor but not cognitive functions in Niemann-Pick Type C1 mice. Neuroscience 2013; 252:201-11. [PMID: 23948640 DOI: 10.1016/j.neuroscience.2013.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 12/21/2022]
Abstract
Niemann-Pick Type C1 (NPC1) is an autosomal recessive disorder characterized by the accumulation of cholesterol and glycosphingolipids. Combination-treatment utilizing cyclodextrin, allopregnanolone and miglustat (CYCLO/ALLO/miglustat) can ameliorate NPC1 disease in a mutant mouse model. The present study was designed to add behavioral analysis in NPC1 mutant mice upon CYCLO/ALLO/miglustat therapy. NPC1 mutant (BALB/cJ NPC1NIH) and control mice were used. For the combination treatment mice were injected with CYCLO/ALLO weekly, starting at P7. The miglustat injection was performed daily from P10 till P23. Starting at P23, miglustat was added to the powdered chow. For the sham treatment of control and mutant mice the same schedule was used with 0.9% NaCl injection. Locomotor activity was assessed in open field, elevated plus maze and accelerod tests. For assessment of spatial learning and memory the Morris water maze test was conducted. Electron microscopy has been performed to support the behavioral data. The sham-treated mutant mice exhibited motor impairments in all performed tests. In the water maze the sham-treated mutants exhibited impairment in remembering the location of the hidden platform. CYCLO/ALLO/miglustat treatment positively influenced motor dysfunction: total distance and number of visits significantly increased, and accelerod performance improved. The spatial learning, however, did not benefit from therapy. At the morphological level, an excessive accumulation of electron-dense material was seen in the cerebellar Purkinje cells of mutant mice. A regression of these autophagosomal inclusions was seen upon therapy. CYCLO/ALLO/miglustat therapy ameliorates motor but not cognitive deficits in NPC1 mutant mice, suggesting unequal vulnerability of different brain areas to the treatment.
Collapse
Affiliation(s)
- M Hovakimyan
- Institute of Anatomy, University of Rostock, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sun C, Shao J, Su L, Zhao J, Bi J, Yang S, Zhang S, Gao J, Miao J. Cholinergic Neuron-Like Cells Derived from Bone Marrow Stromal Cells Induced by Tricyclodecane-9-yl-Xanthogenate Promote Functional Recovery and Neural Protection after Spinal Cord Injury. Cell Transplant 2013; 22:961-75. [PMID: 23031841 DOI: 10.3727/096368912x657413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The rate of neuronal differentiation of bone marrow stromal cells (BMSCs) in vivo is very low; therefore, it is necessary to elevate the number of BMSC-derived neurons to cure neurodegenerative diseases. We previously reported that tricyclodecane-9-yl-xanthogenate (D609), an inhibitor of phosphatidylcholine-specific phospholipase C (PC-PLC), induced BMSCs to differentiate into neuron-like cells in vitro. However, the neuronal type is not clear, and it is still unknown whether these neuron-like cells possess physiological properties of functional neurons and whether they can contribute to the recovery of neuron dysfunction. To answer these questions, we investigated their characteristics by detecting neuronal function-related neurotransmitters and calcium image. The results showed that these cells exhibited functional cholinergic neurons in vitro. Transplantation of these cholinergic neuron-like cells promoted the recovery of spinal cord-injured mice, and they were more effective than BMSCs. The number of cholinergic neurons was increased after injection with BMSC-derived cholinergic neuron-like cells, indicating their high differentiation rate in vivo. Moreover, the proportion of cholinergic neurons in host cells and secretion of acetylcholine were increased, and preservation of neurofilament was also observed in the lesion of mice implanted with BMSC-derived neurons, suggesting the neuronal protection of BMSC-derived neurons. Our findings provide both a simple method to induce the differentiation of BMSCs into cholinergic neuron-like cells and a putative strategy for the therapy of spinal cord injuries.
Collapse
Affiliation(s)
- Chunhui Sun
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
| | - Jing Shao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
| | - Le Su
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Qilu Hospital, Jinan, China
| | - Jing Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Qilu Hospital, Jinan, China
| | - Jianzhong Bi
- Department of Neural Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Shaonan Yang
- Department of Neural Medicine, The Second Hospital of Shandong University, Jinan, China
| | - Shangli Zhang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Qilu Hospital, Jinan, China
| | - Jiangang Gao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
| | - Junying Miao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University, Qilu Hospital, Jinan, China
| |
Collapse
|
46
|
Liu J, Chen J, Liu B, Yang C, Xie D, Zheng X, Xu S, Chen T, Wang L, Zhang Z, Bai X, Jin D. Acellular spinal cord scaffold seeded with mesenchymal stem cells promotes long-distance axon regeneration and functional recovery in spinal cord injured rats. J Neurol Sci 2013; 325:127-36. [PMID: 23317924 DOI: 10.1016/j.jns.2012.11.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2012] [Revised: 10/23/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023]
Abstract
The stem cell-based experimental therapies are partially successful for the recovery of spinal cord injury (SCI). Recently, acellular spinal cord (ASC) scaffolds which mimic native extracellular matrix (ECM) have been successfully prepared. This study aimed at investigating whether the spinal cord lesion gap could be bridged by implantation of bionic-designed ASC scaffold alone and seeded with human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) respectively, and their effects on functional improvement. A laterally hemisected SCI lesion was performed in adult Sprague-Dawley (SD) rats (n=36) and ASC scaffolds seeded with or without hUCB-MSCs were implanted into the lesion immediately. All rats were behaviorally tested using the Basso-Beattie-Bresnahan (BBB) test once a week for 8weeks. Behavioral analysis showed that there was significant locomotor recovery improvement in combined treatment group (ASC scaffold and ASC scaffold+hUCB-MSCs) as compared with the SCI only group (p<0.01). 5-Bromodeoxyuridine (Brdu)-labeled hUCB-MSCs could also be observed in the implanted ACS scaffold two weeks after implantation. Moreover, host neural cells (mainly oligodendrocytes) were able to migrate into the graft. Biotin-dextran-amine (BDA) tracing test demonstrated that myelinated axons successfully grew into the graft and subsequently promoted axonal regeneration at lesion sites. This study provides evidence for the first time that ASC scaffold seeded with hUCB-MSCs is able to bridge a spinal cord cavity and promote long-distance axon regeneration and functional recovery in SCI rats.
Collapse
Affiliation(s)
- Jia Liu
- Department of Orthopedics, the Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Roh DH, Seo MS, Choi HS, Park SB, Han HJ, Beitz AJ, Kang KS, Lee JH. Transplantation of human umbilical cord blood or amniotic epithelial stem cells alleviates mechanical allodynia after spinal cord injury in rats. Cell Transplant 2013; 22:1577-90. [PMID: 23294734 DOI: 10.3727/096368912x659907] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Stem cell therapy is a potential treatment for spinal cord injury (SCI), and a variety of different stem cell types have been grafted into humans suffering from spinal cord trauma or into animal models of spinal injury. Although several studies have reported functional motor improvement after transplantation of stem cells into injured spinal cord, the benefit of these cells for treating SCI-induced neuropathic pain is not clear. In this study, we investigated the therapeutic effect of transplanting human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) or amniotic epithelial stem cells (hAESCs) on SCI-induced mechanical allodynia (MA) and thermal hyperalgesia (TH) in T13 spinal cord hemisected rats. Two weeks after SCI, hUCB-MSCs or hAESCs were transplanted around the spinal cord lesion site, and behavioral tests were performed to evaluate changes in SCI-induced MA and TH. Immunohistochemical and Western blot analyses were also performed to evaluate possible therapeutic effects on SCI-induced inflammation and the nociceptive-related phosphorylation of the NMDA NR1 receptor subunit. While transplantation of hUCB-MSCs showed a tendency to reduce MA, transplantation of hAESCs significantly reduced MA. Neither hUCB-MSC nor hAESC transplantation had any effect on SCI-induced TH. Transplantation of hAESCs also significantly reduced the SCI-induced increase in NMDA receptor NR1 subunit phosphorylation (pNR1) expression in the spinal cord. Both hUCB-MSCs and hAESCs reduced the SCI-induced increase in spinal cord expression of the microglial marker, F4/80, but not the increased expression of GFAP or iNOS. Taken together, these findings demonstrate that the transplantation of hAESCs into the injured spinal cord can suppress mechanical allodynia, and this effect seems to be closely associated with the modulation of spinal cord microglia activity and NR1 phosphorylation.
Collapse
Affiliation(s)
- Dae-Hyun Roh
- Department of Maxillofacial Tissue Regeneration, School of Dentistry, Kyung Hee University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim J, Jeon YJ, Kim HE, Shin JM, Chung HM, Chae JI. Comparative proteomic analysis of endothelial cells progenitor cells derived from cord blood- and peripheral blood for cell therapy. Biomaterials 2012; 34:1669-85. [PMID: 23218840 DOI: 10.1016/j.biomaterials.2012.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/10/2012] [Indexed: 11/29/2022]
Abstract
Vasculopathy due to ischemia in damaged tissues is a major cause of morbidity and mortality. To treat these conditions, endothelial progenitor cells (EPCs) from various sources, such as umbilical cord or peripheral blood, have been the focus of the regenerative medicine field due to their proliferative and vasculogenic activities. However, the fundamental, molecular-level differences between EPCs obtained from different cellular sources have rarely been studied. In this study, we established endothelial progenitor cells derived from cord blood- and peripheral blood (CB- and PB-EPCs) and investigated their fundamental differences at the cellular and molecular levels through a combination of stem cell biology techniques and proteomic analysis. Our results suggest that specifically up-regulated factors such as STMIN 1, CFL 1, PARK 7, NME 1, GLO 1, HSP 27 and PRDX 2 in CB-EPCs as key elements that could be functionally active in ischemic regions. We also discussed functional behaviors important for inducing and maintaining long-lasting blood vessels under ischemic conditions. As a result, CB-EPCs retained a higher anti-oxidant and migration ability than PB-EPCs in vitro. Furthermore, CB-EPCs retained a higher therapeutic efficacy than PB-EPCs in a hindlimb ischemic disease model. The up-regulated expression pattern of STMIN 1, CFL 1, PARK 7, NME 1, GLO 1, HSP 27 and PRDX 2 was confirmed under several conditions in vitro and in vivo, indicating that the up-regulation of these molecules in CB-EPCs may be critical to the mechanism of healing in ischemic conditions and that CB-EPCs may be more appropriate for inducing neo-vessels. Thus, these results may aid in predetermining which cell sources will be of value for cell-based therapies of pathological conditions and identify several candidate molecules that may be involved in the therapeutic mechanism for ischemia.
Collapse
Affiliation(s)
- Jumi Kim
- CHA Bio & Diostech Co., Ltd., 606-16 Yeoksam 1 dong, Gangnam gu, Seoul 135-907, Republic of Korea
| | | | | | | | | | | |
Collapse
|
49
|
Seo Y, Yang SR, Kim HS, Yu KR, Shin Y, Kang SK, Kang KS. JNK Activation by Up-Regulation of iNOS on Cholesterol Accumulation Limits Neurogenesis and Induces Region-Specific DNA Damage Responses in the Subventricular Zone of NPC Mice. Antioxid Redox Signal 2012:120410100912001. [PMID: 22384812 DOI: 10.1089/ars.2011.4301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Abstract Aims: We explore the region-specific impact of nitric oxide (NO) on adult neural stem cell (aNSC) niches with regard to neurogenesis and NSC damage and investigate the underlying mechanisms in Niemann-Pick disease type C (NPC) mice. Results: Among the two anatomical stem-cell niches of the brain, subventricular zone (SVZ)-derived aNSCs enhanced c-Jun N-terminal kinase (JNK) activity because of excessive NO production by the cholesterol accumulation. Activated JNK interacts with γH2AX, a marker for DNA damage; however, almost none of the aNSCs in the dentate gyrus (DG) showed either JNK signaling activation or abundant DNA damage. SVZ-derived aNSCs were protected from DNA damage by the treatment of Nω-nitro-L-arginine methyl ester (L-NAME), a NO synthase (NOS) inhibitor, both in vitro and in vivo. We also observed that U18666A, an inducer of cholesterol accumulation, increased inducible NOS expression, JNK activation, and DNA damage in the wild type (WT)-aNSCs. Interestingly, we found that endogenous cholesterol efflux transporters and their regulator were less activated in the SVZ than in the DG, in both WT and NPC mice. This result explains the high vulnerability of SVZ-derived aNSCs to the cholesterol imbalance as observed in NPC mice. Innovation and Conclusion: In this study, we demonstrated that the SVZ-derived aNSCs might be major targets of NPC. Significantly, aNSCs showed different responses depending on their anatomical origins due to dissimilarities in their cholesterol transporting system and NO-dependent JNK activation. These findings can contribute to the understanding of the region-specific nature of the two SVZ and DG neurogenic niches. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Yoojin Seo
- 1 Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University , Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Dalous J, Larghero J, Baud O. Transplantation of umbilical cord-derived mesenchymal stem cells as a novel strategy to protect the central nervous system: technical aspects, preclinical studies, and clinical perspectives. Pediatr Res 2012; 71:482-90. [PMID: 22430384 DOI: 10.1038/pr.2011.67] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The prevention of perinatal neurological disabilities remains a major challenge for public health, and no neuroprotective treatment to date has proven clinically useful in reducing the lesions leading to these disabilities. Efforts are, therefore, urgently needed to test other neuroprotective strategies including cell therapies. Although stem cells have raised great hopes as an inexhaustible source of therapeutic products that could be used for neuroprotection and neuroregeneration in disorders affecting the brain and spinal cord, certain sources of stem cells are associated with potential ethical issues. The human umbilical cord (hUC) is a rich source of stem and progenitor cells including mesenchymal stem cells (MSCs) derived either from the cord or from cord blood. hUC MSCs (hUC-MSCs) have several advantages as compared to other types and sources of stem cells. In this review, we will summarize the most recent findings regarding the technical aspects and the preclinical investigation of these promising cells in neuroprotection and neuroregeneration, and their potential use in the developing human brain. However, extensive studies are needed to optimize the administration protocol, safety parameters, and potential preinjection cell manipulations before designing a controlled trial in human neonates.
Collapse
Affiliation(s)
- Jérémie Dalous
- INSERM UMR 676, Université Paris Diderot, Hôpital Robert Debré, APHP, Paris, France
| | | | | |
Collapse
|