1
|
Qu Y, Wang Z, Dong L, Zhang D, Shang F, Li A, Gao Y, Bai Q, Liu D, Xie X, Ming L. Natural small molecules synergize mesenchymal stem cells for injury repair in vital organs: a comprehensive review. Stem Cell Res Ther 2024; 15:243. [PMID: 39113141 PMCID: PMC11304890 DOI: 10.1186/s13287-024-03856-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Mesenchymal stem cells (MSCs) therapy is a highly researched treatment that has the potential to promote immunomodulation and anti-inflammatory, anti-apoptotic, and antimicrobial activities. It is thought that it can enhance internal organ function, reverse tissue remodeling, and achieve significant organ repair and regeneration. However, the limited infusion, survival, and engraftment of transplanted MSCs diminish the effectiveness of MSCs-based therapy. Consequently, various preconditioning methods have emerged as strategies for enhancing the therapeutic effects of MSCs and achieving better clinical outcomes. In particular, the use of natural small molecule compounds (NSMs) as a pretreatment strategy is discussed in this narrative review, with a focus on their roles in regulating MSCs for injury repair in vital internal organs. Additionally, the discussion focuses on the future directions and challenges of transforming mesenchymal stem cell research into clinical applications.
Collapse
Affiliation(s)
- Yanling Qu
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Zhe Wang
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Lingjuan Dong
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Dan Zhang
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Fengqing Shang
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, China
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510000, China
| | - Afeng Li
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Yanni Gao
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Qinhua Bai
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Dan Liu
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China
| | - Xiaodong Xie
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| | - Leiguo Ming
- Shaanxi Zhonghong, Institute of Regenerative Medicine, Xi'an, 710003, Shaanxi Province, China.
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
2
|
Hori H, Sakai K, Ohashi A, Nakai S. Chitin powder enhances growth factor production and therapeutic effects of mesenchymal stem cells in a chronic kidney disease rat model. J Artif Organs 2023; 26:203-211. [PMID: 35976577 DOI: 10.1007/s10047-022-01346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/06/2022] [Indexed: 10/15/2022]
Abstract
Previously, we fabricated a device with polylactic acid nonwoven filters and mesenchymal stem cells (MSCs), which effectively reduced urinary protein levels in a rat model of chronic kidney disease (CKD) but could not suppress CKD progression. Therefore, to improve the therapeutic effects of MSCs, in this study, we analyzed the ability of rat adipose tissue-derived MSCs (ADSCs) in contact with chitin nonwoven filters or chitin powder to produce growth factors and examined their therapeutic effect in an adriamycin (ADR)-induced CKD rat model. Hepatocyte growth factor (HGF) and vascular endothelial growth factor (VEGF) production was significantly enhanced by ADSCs cultured in a medium containing chitin powder (C-ADSCs) compared with that by ADSCs cultured in a standard medium without chitin (N-ADSCs). However, the production of HGF and VEGF by ADSCs on chitin nonwoven filters was not significantly enhanced compared with that by the control. Intravenous C-ADSC injection significantly increased podocin expression and improved proteinuria compared with those in saline-treated CKD rats; however, no such improvements were observed in the N-ADSC-treated group. These results showed that ADSCs cultured in a medium supplemented with chitin powder suppressed proteinuria via enhanced HGF and VEGF production in ADR-induced CKD rats to mitigate podocyte damage, offering a new strategy to reduce the dose of MSC therapy for safe and effective treatment of kidney disease.
Collapse
Affiliation(s)
- Hideo Hori
- Faculty of Medical Technology, School of Medical Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| | - Kazuyoshi Sakai
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Atsushi Ohashi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Shigeru Nakai
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
3
|
Stem cell-nanomedicine system as a theranostic bio-gadolinium agent for targeted neutron capture cancer therapy. Nat Commun 2023; 14:285. [PMID: 36650171 PMCID: PMC9845336 DOI: 10.1038/s41467-023-35935-0] [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: 09/16/2021] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
The potential clinical application of gadolinium-neutron capture therapy (Gd-NCT) for glioblastoma multiforme (GBM) treatment has been compromised by the fast clearance and nonspecific biodistribution of gadolinium-based agents. We have developed a stem cell-nanoparticle system (SNS) to actively target GBM for advanced Gd-NCT by magnetizing umbilical cord mesenchymal stem cells (UMSCs) using gadodiamide-concealed magnetic nanoparticles (Gd-FPFNP). Nanoformulated gadodiamide shielded by a dense surface composed of fucoidan and polyvinyl alcohol demonstrates enhanced cellular association and biocompatibility in UMSCs. The SNS preserves the ability of UMSCs to actively penetrate the blood brain barrier and home to GBM and, when magnetically navigates by an external magnetic field, an 8-fold increase in tumor-to-blood ratio is achieved compared with clinical data. In an orthotopic GBM-bearing rat model, using a single dose of irradiation and an ultra-low gadolinium dose (200 μg kg-1), SNS significantly attenuates GBM progression without inducing safety issues, prolonging median survival 2.5-fold compared to free gadodiamide. The SNS is a cell-based delivery system that integrates the strengths of cell therapy and nanotechnology, which provides an alternative strategy for the treatment of brain diseases.
Collapse
|
4
|
Li C, Cui Z, Deng S, Chen P, Li X, Yang H. The potential of plant extracts in cell therapy. STEM CELL RESEARCH & THERAPY 2022; 13:472. [PMID: 36104798 PMCID: PMC9476258 DOI: 10.1186/s13287-022-03152-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 05/23/2022] [Indexed: 11/10/2022]
Abstract
Cell therapy is the frontier technology of biotechnology innovation and the most promising method for the treatment of refractory diseases such as tumours. However, cell therapy has disadvantages, such as toxicity and poor therapeutic effects. Plant extracts are natural, widely available, and contain active small molecule ingredients that are widely used in the treatment of various diseases. By studying the effect of plant extracts on cell therapy, active plant extracts that have positive significance in cell therapy can be discovered, and certain contributions to solving the current problems of attenuation and adjuvant therapy in cell therapy can be made. Therefore, this article reviews the currently reported effects of plant extracts in stem cell therapy and immune cell therapy, especially the effects of plant extracts on the proliferation and differentiation of mesenchymal stem cells and nerve stem cells and the potential role of plant extracts in chimeric antigen receptor T-cell immunotherapy (CAR-T) and T-cell receptor modified T-cell immunotherapy (TCR-T), in the hope of encouraging further research and clinical application of plant extracts in cell therapy.
Collapse
|
5
|
Kim M, Hayashi M, Yu B, Lee TK, Kim RH, Jo DW. Effects of Fucoidan Powder Combined with Mineral Trioxide Aggregate as a Direct Pulp-Capping Material. Polymers (Basel) 2022; 14:polym14122315. [PMID: 35745892 PMCID: PMC9228542 DOI: 10.3390/polym14122315] [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] [Received: 05/14/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
The development of direct pulp-capping materials with favorable biological and structural properties is an important goal in restorative dentistry. Fucoidan is a sulfated, fucose-containing polysaccharide obtained from brown seaweed, with a wide range of applications; however, its use as a direct pulp-capping material has not been examined. This study aimed to evaluate the mechanical, physical, and biological effects of fucoidan combined with conventional mineral trioxide aggregate (MTA) for direct pulp capping. The capping materials were created using Portland cement (80 wt%) and zirconium oxide (20 wt%) as base components, compared with base components plus 5 wt% fucoidan (PZF5) and base components plus 10 wt% fucoidan (PZF10). The initial and final setting time, compressive strength, chemical components, cell viability, adhesion, migration, osteogenesis, and gene expression were analyzed. Fucoidan significantly reduced the initial and final setting time, regardless of quantity. However, the compressive strength was lower for PZF5. Sulfur levels increased with fucoidan. The biological activity improved, especially in the PZF5 group. Cell migration, Alizarin Red S staining, and alkaline phosphatase activity were upregulated in the PZF5 group. Fucoidan is a useful regenerative additive for conventional pulp-capping materials because it reduces the setting time and improves cell migration and osteogenic ability.
Collapse
Affiliation(s)
- Mijoo Kim
- UCLA School of Dentistry Restorative Materials and Applied Dental Research Laboratory, Los Angeles, CA 90095, USA; (M.K.); (M.H.); (B.Y.); (T.K.L.); (R.H.K.)
- Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Marc Hayashi
- UCLA School of Dentistry Restorative Materials and Applied Dental Research Laboratory, Los Angeles, CA 90095, USA; (M.K.); (M.H.); (B.Y.); (T.K.L.); (R.H.K.)
- Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Bo Yu
- UCLA School of Dentistry Restorative Materials and Applied Dental Research Laboratory, Los Angeles, CA 90095, USA; (M.K.); (M.H.); (B.Y.); (T.K.L.); (R.H.K.)
- Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Thomas K. Lee
- UCLA School of Dentistry Restorative Materials and Applied Dental Research Laboratory, Los Angeles, CA 90095, USA; (M.K.); (M.H.); (B.Y.); (T.K.L.); (R.H.K.)
- Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Reuben H. Kim
- UCLA School of Dentistry Restorative Materials and Applied Dental Research Laboratory, Los Angeles, CA 90095, USA; (M.K.); (M.H.); (B.Y.); (T.K.L.); (R.H.K.)
- Section of Restorative Dentistry, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Deuk-Won Jo
- Section of Dentistry, Department of Prosthodontics, Seoul National University Bundang Hospital, Seongnam 13620, Korea
- Correspondence: ; Tel.: +82-31-787-7548
| |
Collapse
|
6
|
Wang J, Lin Y, Chen X, Liu Y, Zhou T. Mesenchymal stem cells: A new therapeutic tool for chronic kidney disease. Front Cell Dev Biol 2022; 10:910592. [PMID: 36268508 PMCID: PMC9577598 DOI: 10.3389/fcell.2022.910592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/12/2022] [Indexed: 02/05/2023] Open
Abstract
Chronic kidney disease (CKD) has a major impact on public health, which could progress to end-stage kidney disease (ESRD) and consume many medical resources. Currently, the treatment for CKD has many flaws, so more effective treatment tools are urgently required for CKD. Mesenchymal stem cells (MSCs) are primitive cells with self-renewal and proliferation capacity and differentiation potential. Extensive preclinical and clinical data has shown that cell-based therapies using MSCs can modulate immunity, inhibit inflammatory factors, and improve renal function in CKD, suggesting that MSCs have the potential to be a new, effective therapeutic tool for CKD. In this review, we will describe different kinds of MSCs and MSCs products for the treatment of CKD in experimental models and clinical trials, potential signaling pathways, therapeutic efficacy, and critical issues that need to be addressed before therapeutic application in humans.
Collapse
Affiliation(s)
- Jiali Wang
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yongda Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Xiutian Chen
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Yiping Liu
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, Shantou, China
| |
Collapse
|
7
|
Begum R, Howlader S, Mamun-Or-Rashid ANM, Rafiquzzaman SM, Ashraf GM, Albadrani GM, Sayed AA, Peluso I, Abdel-Daim MM, Uddin MS. Antioxidant and Signal-Modulating Effects of Brown Seaweed-Derived Compounds against Oxidative Stress-Associated Pathology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9974890. [PMID: 34336128 PMCID: PMC8289617 DOI: 10.1155/2021/9974890] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 11/25/2022]
Abstract
The biological and therapeutic properties of seaweeds have already been well known. Several studies showed that among the various natural marine sources of antioxidants, seaweeds have become a potential source of antioxidants because of their bioactive compounds. Most of the metabolic diseases are caused by oxidative stress. It is very well known that antioxidants have a pivotal role in the treatment of those diseases. Recent researches have revealed the potential activity of seaweeds as complementary medicine, which have therapeutic properties for health and disease management. Among the seaweeds, brown seaweeds (Phaeophyta) and their derived bioactive substances showed excellent antioxidant properties than other seaweeds. This review focuses on brown seaweeds and their derived major bioactive compounds such as sulfated polysaccharide, polyphenol, carotenoid, and sterol antioxidant effects and molecular mechanisms in the case of the oxidative stress-originated disease. Antioxidants have a potential role in the modification of stress-induced signaling pathways along with the activation of the oxidative defensive pathways. This review would help to provide the basis for further studies to researchers on the potential antioxidant role in the field of medical health care and future drug development.
Collapse
Affiliation(s)
- Rahima Begum
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-do, 26426, Republic of Korea
| | - Saurav Howlader
- Department of Pharmacology and Pharmaco Genomics Research Centre (PGRC), Inje University College of Medicine, Busan, Republic of Korea
| | - A. N. M. Mamun-Or-Rashid
- Anti-Aging Medical Research Center and Glycative Stress Research Center, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto 610-0394, Japan
| | - S. M. Rafiquzzaman
- Department of Fisheries Biology & Aquatic Environment, Bangabandhu Sheikh Mujibur Rahman Agricultural University (BSMRAU), Gazipur 1706, Bangladesh
| | - Ghulam Md Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Amany A. Sayed
- Zoology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), 00142 Rome, Italy
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| |
Collapse
|
8
|
Baek SJ, Hammock BD, Hwang IK, Li Q, Moustaid-Moussa N, Park Y, Safe S, Suh N, Yi SS, Zeldin DC, Zhong Q, Bradbury JA, Edin ML, Graves JP, Jung HY, Jung YH, Kim MB, Kim W, Lee J, Li H, Moon JS, Yoo ID, Yue Y, Lee JY, Han HJ. Natural Products in the Prevention of Metabolic Diseases: Lessons Learned from the 20th KAST Frontier Scientists Workshop. Nutrients 2021; 13:1881. [PMID: 34072678 PMCID: PMC8227583 DOI: 10.3390/nu13061881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
The incidence of metabolic and chronic diseases including cancer, obesity, inflammation-related diseases sharply increased in the 21st century. Major underlying causes for these diseases are inflammation and oxidative stress. Accordingly, natural products and their bioactive components are obvious therapeutic agents for these diseases, given their antioxidant and anti-inflammatory properties. Research in this area has been significantly expanded to include chemical identification of these compounds using advanced analytical techniques, determining their mechanism of action, food fortification and supplement development, and enhancing their bioavailability and bioactivity using nanotechnology. These timely topics were discussed at the 20th Frontier Scientists Workshop sponsored by the Korean Academy of Science and Technology, held at the University of Hawaii at Manoa on 23 November 2019. Scientists from South Korea and the U.S. shared their recent research under the overarching theme of Bioactive Compounds, Nanoparticles, and Disease Prevention. This review summarizes presentations at the workshop to provide current knowledge of the role of natural products in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Seung J. Baek
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Bruce D. Hammock
- Department of Entomology, University of California, Davis, CA 95616, USA;
| | - In-Koo Hwang
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Qingxiao Li
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI 96822, USA;
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences & Obesity Research Institute, Texas Tech University, Lubbock, TX 79409, USA;
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Stephen Safe
- Department of Biochemistry & Biophysics, Texas A & M University, College Station, TX 77843, USA;
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
| | - Sun-Shin Yi
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Darryl C. Zeldin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Qixin Zhong
- Department of Food Sciences, University of Tennessee, Knoxville, TN 37996, USA;
| | - Jennifer Alyce Bradbury
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Matthew L. Edin
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Joan P. Graves
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Hyo-Young Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Young-Hyun Jung
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Mi-Bo Kim
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Woosuk Kim
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Jaehak Lee
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| | - Hong Li
- National Institutes of Environmental Health, National Institutes of Health, Research Triangle Park, NC 27709, USA; (D.C.Z.); (J.A.B.); (M.L.E.); (J.P.G.); (H.L.)
| | - Jong-Seok Moon
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Ik-Dong Yoo
- Department of Medical Sciences, Soonchunhyang University, Asan 31538, Korea; (S.-S.Y.); (J.-S.M.); (I.-D.Y.)
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA; (Y.P.); (Y.Y.)
| | - Ji-Young Lee
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Ho-Jae Han
- College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.J.B.); (I.-K.H.); (H.-Y.J.); (Y.-H.J.); (W.K.); (J.L.)
| |
Collapse
|
9
|
Chen YC, Cheng CY, Liu CT, Sue YM, Chen TH, Hsu YH, Huang NJ, Chen CH. Combined protective effects of oligo-fucoidan, fucoxanthin, and L-carnitine on the kidneys of chronic kidney disease mice. Eur J Pharmacol 2021; 892:173708. [PMID: 33152336 DOI: 10.1016/j.ejphar.2020.173708] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is a common global progressive disease, but there are no ideal drugs for the treatment. Fucoidan and fucoxanthin, and L-carnitine are one of the very few natural products that have a therapeutic effect on CKD in animal experiments. However, the combined effects of these compounds on CKD are unknown. We established a mouse CKD model by right nephrectomy with transient ischemic injury to the left kidney. Oligo-fucoidan and fucoidan were extracted from Laminaria japonica. We fed CKD mice with the two compounds and L-carnitine to evaluate the combined effects on CKD. Oligo-fucoidan and fucoidan inhibited renal fibrosis and reduced serum creatine in CKD mice to a greater extent than any single compound. L-carnitine had no measurable effect on renal fibrosis but promoted the protective effect of the mixture of oligo-fucoidan and fucoidan on renal function in CKD mice. In the two-month safety test, the combined mixture further improved renal function and did not elevate serum aspartate aminotransferase and alanine aminotransferase levels in CKD mice. Furthermore, the weights of CKD mice treated with the combination increased to the normal level. We also found that all oligo-fucoidan, fucoxanthin, and L-carnitine inhibit H2O2-induced apoptosis and activated Akt in rat renal tubular cells. Our results confirm that oligo-fucoidan, fucoxanthin, and L-carnitine have a combined protective effect on the kidneys. The combined mixture may be beneficial for CKD patients.
Collapse
Affiliation(s)
- Yen-Cheng Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Chung-Yi Cheng
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Chung-Te Liu
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Yuh-Mou Sue
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Tso-Hsiao Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Yung-Ho Hsu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan
| | - Nai-Jen Huang
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Urology and Kidney, Taipei, Taiwan.
| |
Collapse
|
10
|
Zhang Y, Du H, Yu X, Zhu J. Fucoidan attenuates hyperoxia-induced lung injury in newborn rats by mediating lung fibroblasts differentiate into myofibroblasts. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1501. [PMID: 33313246 PMCID: PMC7729344 DOI: 10.21037/atm-20-6601] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Hyperoxia-induced lung injury is one of the most common and frequent diseases in premature infants and may develop into bronchopulmonary dysplasia (BPD). Fucoidan, extracted from brown seaweed and brown algae, has anti-apoptosis, antioxidative and anti-fibrosis effects. This study aimed to explore whether fucoidan could alleviate hyperoxia-induced lung injury in newborn rats. Methods Lung wet-weight/dry-weight (W/D) ratio, total protein (TP) content, total cell counts, and lactate dehydrogenase (LDH) levels are used to evaluate lung injury. Masson staining is used to evaluate lung fibrotic. Tunnel assay and Hoechst 33258 assay were used to evaluate apoptosis. The levels of serum superoxide dismutase (SOD), malondialdehyde (MDA), and glutathione (GSH) were measured using ELISA to assess oxidative stress. Western blot assay was used to detect apoptosis-related proteins Bcl-1, Bax, and myofibroblast proteins α-SMA. Results The data indicating fucoidan treatment remarkably reduces the lung W/D ratio and TP content, total cell counts, and LDH levels in bronchoalveolar lavage fluid (BALF). Also, fucoidan treatment significantly inhibited cell apoptosis with the elevated expression of Bcl-2/Bax in cultured lung fibroblasts. Moreover, treatment with fucoidan suppressed the levels of MDA significantly and elevated the level of SOD and GSH, showing that oxidative stress was restrained by fucoidan. Furthermore, the decreased expression levels of α-SMA and collagen I was detected in fibroblast treated with fucoidan. Conclusions These data suggest fucoidan may protect the lung from hyperoxia via suppressing cell apoptosis, mitigating oxidative stress, and inhibiting lung fibroblasts from differentiating into myofibroblasts.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Hengjian Du
- Department of Geriatric Infectious Diseases, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Xuelian Yu
- Department of Geriatric Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| | - Jiang Zhu
- Department of Respiratory and Critical Care Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University Hospital of Electronic Science & Technology, Chengdu, China
| |
Collapse
|
11
|
Nunes C, Coimbra MA. The Potential of Fucose-Containing Sulfated Polysaccharides As Scaffolds for Biomedical Applications. Curr Med Chem 2019; 26:6399-6411. [PMID: 30543164 DOI: 10.2174/0929867326666181213093718] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 12/13/2022]
Abstract
Marine environments have a high quantity and diversity of sulfated polysaccharides. In coastal regions brown algae are the most abundant biomass producers and their cell walls have fucosecontaining sulfated polysaccharides (FCSP), known as fucans and/or fucoidans. These sulfated compounds have been widely researched for their biomedical properties, namely the immunomodulatory, haemostasis, pathogen inhibition, anti-inflammatory capacity, and antitumoral. These activities are probably due to their ability to mimic the carbohydrate moieties of mammalian glycosaminoglycans. Therefore, the FCSP are interesting compounds for application in health-related subjects, mainly for developing scaffolds for delivery systems or tissue regeneration. FCSP showed potential for these applications also due to their ability to form stable 3D structures with other polymers able to entrap therapeutic agents or cell and growth factors, besides their biocompatibility and biodegradability. However, for the clinical use of these biopolymers well-defined reproducible molecules are required in order to accurately establish relationships between structural features and human health applications.
Collapse
Affiliation(s)
- Cláudia Nunes
- CICECO-Aveiro Institute of Materials, University of Aveiro, 3810-193 Aveiro, Portugal.,QOPNA/LAQVREQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Manuel A Coimbra
- QOPNA/LAQVREQUIMTE, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
12
|
Li Y, Zhao W, Wang L, Chen Y, Zhang H, Wang T, Yang X, Xing F, Yan J, Fang X. Protective Effects of Fucoidan against Hydrogen Peroxide-Induced Oxidative Damage in Porcine Intestinal Epithelial Cells. Animals (Basel) 2019; 9:ani9121108. [PMID: 31835456 PMCID: PMC6940796 DOI: 10.3390/ani9121108] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/30/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
Simple Summary High levels of production in intensive farming systems make domestic animals like piglets particularly susceptible to oxidative stress, which is detrimental to intestinal homeostasis and function. It is of paramount importance to identify effective and reliable nutrients to counteract oxidative damage to the porcine intestinal epithelium, especially with the recent phasing out of the use of antibiotics in China. This study indicates that fucoidan could ameliorate hydrogen peroxide-induced oxidative stress in porcine intestinal epithelial cells, primarily owing to the action of fucoidan to facilitate nuclear factor-erythroid 2-related factor-2 signals and cellular antioxidant responses. These findings may provide useful implications for practical swine production. Abstract This study was conducted to evaluate the effectiveness of fucoidan in ameliorating hydrogen peroxide (H2O2)-induced oxidative stress to porcine intestinal epithelial cell line (IPEC-1). The cell viability test was initially performed to screen out appropriate concentrations of H2O2 and fucoidan. After that, cells were exposed to H2O2 in the presence or absence of pre-incubation with fucoidan. Hydrogen peroxide increased the apoptotic and necrotic rate, boosted reactive oxygen species (ROS) generation, and disturbed the transcriptional expression of genes associated with antioxidant defense and apoptosis in IPEC-1 cells. Pre-incubation with fucoidan inhibited the increases in necrosis and ROS accumulation induced by H2O2. Consistently, in the H2O2-treated IPEC-1 cells, fucoidan normalized the content of reduced glutathione as well as the mRNA abundance of NAD(P)H quinone dehydrogenase 1 and superoxide dismutase 1 while it prevented the overproduction of malondialdehyde. Moreover, H2O2 stimulated the translocation of nuclear factor-erythroid 2-related factor-2 to the nucleus of IPEC-1 cells, but this increase was further promoted by fucoidan pre-treatment. The results suggest that fucoidan is effective in protecting IPEC-1 cells against oxidative damage induced by H2O2, which may help in developing appropriate strategies for maintaining the intestinal health of young piglets.
Collapse
Affiliation(s)
- Yue Li
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Weimin Zhao
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Li Wang
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Yueping Chen
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.C.); (H.Z.); (T.W.)
| | - Hao Zhang
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.C.); (H.Z.); (T.W.)
| | - Tian Wang
- College of Animal Science & Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.C.); (H.Z.); (T.W.)
| | - Xiaoyang Yang
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Fei Xing
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Junshu Yan
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
| | - Xiaomin Fang
- Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; (Y.L.); (W.Z.); (L.W.); (X.Y.); (F.X.); (J.Y.)
- Correspondence: ; Tel.: +86-25-84391941
| |
Collapse
|
13
|
Zhong Q, Wei B, Wang S, Ke S, Chen J, Zhang H, Wang H. The Antioxidant Activity of Polysaccharides Derived from Marine Organisms: An Overview. Mar Drugs 2019; 17:E674. [PMID: 31795427 PMCID: PMC6950075 DOI: 10.3390/md17120674] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/14/2019] [Accepted: 11/22/2019] [Indexed: 12/22/2022] Open
Abstract
Marine-derived antioxidant polysaccharides have aroused extensive attention because of their potential nutritional and therapeutic benefits. However, the comprehensive comparison of identified marine-derived antioxidant polysaccharides is still inaccessible, which would facilitate the discovery of more efficient antioxidants from marine organisms. Thus, this review summarizes the sources, chemical composition, structural characteristics, and antioxidant capacity of marine antioxidant polysaccharides, as well as their protective in vivo effects mediated by antioxidative stress reported in the last few years (2013-2019), and especially highlights the dominant role of marine algae as antioxidant polysaccharide source. In addition, the relationships between the chemical composition and structural characteristics of marine antioxidant polysaccharides with their antioxidant capacity were also discussed. The antioxidant activity was found to be determined by multiple factors, including molecular weight, monosaccharide composition, sulfate position and its degree.
Collapse
Affiliation(s)
- Qiwu Zhong
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| | - Sijia Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
- Center for Human Nutrition, David Geffen School of Medicine, University of California, Rehabilitation Building 32-21, 1000 Veteran Avenue, Los Angeles, CA 90024, USA
| | - Songze Ke
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| | - Jianwei Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| | - Huawei Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China; (Q.Z.); (B.W.); (S.W.); (S.K.); (J.C.); (H.Z.)
| |
Collapse
|
14
|
Deng R, Liu Y, He H, Zhang H, Zhao C, Cui Z, Hong Y, Li X, Lin F, Yuan D, Liang X, Zhang Y. Haemin pre-treatment augments the cardiac protection of mesenchymal stem cells by inhibiting mitochondrial fission and improving survival. J Cell Mol Med 2019; 24:431-440. [PMID: 31660694 PMCID: PMC6933414 DOI: 10.1111/jcmm.14747] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/11/2019] [Accepted: 09/13/2019] [Indexed: 02/06/2023] Open
Abstract
The cardiac protection of mesenchymal stem cell (MSC) transplantation for myocardial infarction (MI) is largely hampered by low cell survival. Haem oxygenase 1 (HO‐1) plays a critical role in regulation of cell survival under many stress conditions. This study aimed to investigate whether pre‐treatment with haemin, a potent HO‐1 inducer, would promote the survival of MSCs under serum deprivation and hypoxia (SD/H) and enhance the cardioprotective effects of MSCs in MI. Bone marrow (BM)‐MSCs were pretreated with or without haemin and then exposed to SD/H. The mitochondrial morphology of MSCs was determined by MitoTracker staining. BM‐MSCs and haemin‐pretreated BM‐MSCs were transplanted into the peri‐infarct region in MI mice. SD/H induced mitochondrial fragmentation, as shown by increased mitochondrial fission and apoptosis of BM‐MSCs. Pre‐treatment with haemin greatly inhibited SD/H‐induced mitochondrial fragmentation and apoptosis of BM‐MSCs. These effects were partially abrogated by knocking down HO‐1. At 4 weeks after transplantation, compared with BM‐MSCs, haemin‐pretreated BM‐MSCs had greatly improved the heart function of mice with MI. These cardioprotective effects were associated with increased cell survival, decreased cardiomyocytes apoptosis and enhanced angiogenesis. Collectively, our study identifies haemin as a regulator of MSC survival and suggests a novel strategy for improving MSC‐based therapy for MI.
Collapse
Affiliation(s)
- Rui Deng
- Department of General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yaming Liu
- School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Haiwei He
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hao Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, China
| | - Chenling Zhao
- Department of Respiratory Medicine, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Zhen Cui
- Department of Radiation Oncology, the First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fang Lin
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dongsheng Yuan
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoting Liang
- Clinical Translational Medical Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuelin Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, China.,Department of Emergency Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
15
|
Melatonin Enhances Mitophagy by Upregulating Expression of Heat Shock 70 kDa Protein 1L in Human Mesenchymal Stem Cells under Oxidative Stress. Int J Mol Sci 2019; 20:ijms20184545. [PMID: 31540288 PMCID: PMC6769944 DOI: 10.3390/ijms20184545] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are a potent source of cell-based regenerative therapeutics used to treat patients with ischemic disease. However, disease-induced oxidative stress disrupts mitochondrial homeostasis in transplanted hMSCs, resulting in hMSC apoptosis and reducing their efficacy post-transplantation. To address this issue, we evaluated the effects of melatonin on cellular defense mechanisms and mitophagy in hMSCs subjected to oxidative stress. H2O2-induced oxidative stress increases the levels of reactive oxygen species and reduces membrane potential in hMSCs, leading to mitochondrial dysfunction and cell death. Oxidative stress also decreases the expression of 70-kDa heat shock protein 1L (HSPA1L), a molecular chaperone that assists in the recruitment of parkin to the autophagosomal mitochondrial membrane. Decreased expression of HSPA1L destabilizes parkin, thereby impairing mitophagy. Our results indicate that treating hMSCs with melatonin significantly inhibited mitochondrial dysfunction induced by oxidative stress, which decreased hMSCs apoptosis. In damaged hMSCs, treatment with melatonin increased the levels of HSPA1L, which bound to parkin. The interaction between HSPA1L and parkin increased membrane potential and levels of oxidative phosphorylation, resulting in enhanced mitophagy. Our results indicate that melatonin increased the expression of HSPA1L, thereby upregulating mitophagy and prolonging cell survival under conditions of oxidative stress. In this study, we have shown that melatonin, a readily available compound, can be used to improve hMSC-based therapies for patients with pathologic conditions involving oxidative stress.
Collapse
|
16
|
Fucoidan Suppresses Mitochondrial Dysfunction and Cell Death against 1-Methyl-4-Phenylpyridinum-Induced Neuronal Cytotoxicity via Regulation of PGC-1α Expression. Mar Drugs 2019; 17:md17090518. [PMID: 31480724 PMCID: PMC6780744 DOI: 10.3390/md17090518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/07/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022] Open
Abstract
Mitochondria are considered to be the powerhouses of cells. They are the most commonly damaged organelles within dopaminergic neurons in patients with Parkinson's disease (PD). Despite the importance of protecting neuronal mitochondria in PD patients, the detailed mechanisms underlying mitochondrial dysfunction during pathogenesis and pathophysiological progression of PD have not yet been elucidated. We investigated the protective action of fucoidan against the detrimental action of 1-methyl-4-phenyl-pyridinium (MPP+), a neurotoxin used to model PD, in the mitochondria of SH-SY5Y neural cells. Fucoidan increased the expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and protected the cells from MPP+-induced apoptosis by upregulating the 5' adenosine monophosphate-activated protein kinase (AMPK)-PGC-1α axis. These effects were blocked by the silencing of the PGC-1α axis. These results indicated that fucoidan protects SH-SY5Y cells from mitochondrial dysfunction and cell death caused by MPP+ treatment, via the AMPK-PGC-1α axis. These findings also suggest that fucoidan could potentially be used as a therapeutic agent for PD.
Collapse
|
17
|
Kim YM, Kim JM, Kim JW, Choi ME, Kim SK, Choi JS. Fucoidan attenuates radioiodine-induced salivary gland dysfunction in mice. BMC Oral Health 2019; 19:198. [PMID: 31470847 PMCID: PMC6716941 DOI: 10.1186/s12903-019-0894-2] [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: 11/19/2018] [Accepted: 08/20/2019] [Indexed: 11/25/2022] Open
Abstract
Background Radioiodine (RI) treatments can destroy the cellular components of salivary glands (SG) and disrupt their function. This study investigated whether fucoidan could attenuate radioiodine-induced SG dysfunction in a mouse model. Methods Female C57BL/6 mice (n = 36) were classified into three groups; i) a normal (control) group, ii) an RI-treated group (0.2 mCi/20 g mouse, administered orally), and iii) a fucoidan and RI-treated group. Mice in each group were classified into three subgroups and sacrificed at 2, 4, and 12 weeks after RI treatment. The measurements of salivary flow rates and lag times and histomorphologic examinations were performed, and apoptotic assays were conducted. Changes in salivary 99mTechnetium (Tc)-pertechnetate parameters using single-photon emission computed tomography were followed. Results Salivary flow rates and lag times in the fucoidan group were improved compared to the RI-treated group. Histologic examinations of SGs in the fucoidan group showed mucin-rich parenchymal areas and reduced periductal fibrosis as compared to the RI-treated group. Moreover, compared with the RI-treated group, fucoidan-treated groups showed evidence of cytoprotection, with a greater number of salivary epithelial cells and myoepithelial cells being observed. Fewer apoptotic cells were observed in the fucoidan group as compared to the RI group. The extent of 99mTc pertechnetate excretion in the fucoidan group was similar to that of the control group. Conclusion Our results demonstrate that fucoidan administration before RI treatment could attenuate RI-induced SG damage and provides a possible candidate for preventing SG damage induced by RI.
Collapse
Affiliation(s)
- Young-Mo Kim
- Translational Research Center, Inha University, Incheon, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Inha University College of Medicine, 27, Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea
| | - Jeong Mi Kim
- Translational Research Center, Inha University, Incheon, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Inha University College of Medicine, 27, Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea
| | - Ji Won Kim
- Translational Research Center, Inha University, Incheon, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Inha University College of Medicine, 27, Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea
| | - Mi Eun Choi
- Translational Research Center, Inha University, Incheon, Republic of Korea.,Department of Otorhinolaryngology-Head and Neck Surgery, Inha University College of Medicine, 27, Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea
| | - Seok-Ki Kim
- Department of Nuclear Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Jeong-Seok Choi
- Translational Research Center, Inha University, Incheon, Republic of Korea. .,Department of Otorhinolaryngology-Head and Neck Surgery, Inha University College of Medicine, 27, Inhang-ro, Jung-gu, Incheon, 22332, Republic of Korea.
| |
Collapse
|
18
|
Asprosin improves the survival of mesenchymal stromal cells in myocardial infarction by inhibiting apoptosis via the activated ERK1/2-SOD2 pathway. Life Sci 2019; 231:116554. [PMID: 31194992 DOI: 10.1016/j.lfs.2019.116554] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022]
Abstract
AIMS Several adipokines have been proven to improve the therapeutic efficacy of mesenchymal stromal cells (MSCs) when used to treat ischemic heart disease. Asprosin (ASP) is a newly-discovered adipokine. ASP might also predict the severity of coronary pathology. We investigated the role of ASP on MSCs and the effects of ASP-pretreated MSCs on myocardial infarction (MI). MAIN METHODS MSCs were labelled with a lentivirus carrying green fluorescent protein (GFP). For in vivo study, after pretreatment with vehicle or ASP, MSCs were injected into infarcted hearts. Cardiac function and fibrosis were then evaluated 4 weeks after the induction of MI and survival of MSCs evaluated after 1 week. MSCs proliferation and migration were investigated after ASP treatment in vitro. MSCs apoptosis induced by hydrogen peroxide (H2O2) was assessed using flow cytometry. KEY FINDINGS Compared to vehicle-pretreated MSCs, ASP-pretreated MSCs significantly improved the left ventricular ejection fraction (LVEF), and inhibited myocardial fibrosis 4 weeks after MI. ASP pretreatment may have promoted homing of transplanted MSCs. In vitro results showed that ASP had no significant effect on MSC proliferation and migration, but protected these cells from H2O2-induced apoptosis. Among 21 molecules associated with antioxidation and cell death, the antioxidant enzyme SOD2 was significantly upregulated by ASP. Furthermore, ASP treatment inhibited H2O2-induced ROS generation and apoptosis via the activated ERK1/2-SOD2 pathway. SIGNIFICANCE This is the first evidence that ASP can regulate MSCs function and enhance MSCs therapy for ischemic heart disease. Furthermore, we demonstrate that ASP protects MSCs from oxidative stress-induced apoptosis via the ERK1/2-SOD2 pathway.
Collapse
|
19
|
Potential and Therapeutic Efficacy of Cell-based Therapy Using Mesenchymal Stem Cells for Acute/chronic Kidney Disease. Int J Mol Sci 2019; 20:ijms20071619. [PMID: 30939749 PMCID: PMC6479813 DOI: 10.3390/ijms20071619] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 12/15/2022] Open
Abstract
Kidney disease can be either acute kidney injury (AKI) or chronic kidney disease (CKD) and it can lead to the development of functional organ failure. Mesenchymal stem cells (MSCs) are derived from a diverse range of human tissues. They are multipotent and have immunomodulatory effects to assist in the recovery from tissue injury and the inhibition of inflammation. Numerous studies have investigated the feasibility, safety, and efficacy of MSC-based therapies for kidney disease. Although the exact mechanism of MSC-based therapy remains uncertain, their therapeutic value in the treatment of a diverse range of kidney diseases has been studied in clinical trials. The use of MSCs is a promising therapeutic strategy for both acute and chronic kidney disease. The mechanism underlying the effects of MSCs on survival rate after transplantation and functional repair of damaged tissue is still ambiguous. The paracrine effects of MSCs on renal recovery, optimization of the microenvironment for cell survival, and control of inflammatory responses are thought to be related to their interaction with the damaged kidney environment. This review discusses recent experimental and clinical findings related to kidney disease, with a focus on the role of MSCs in kidney disease recovery, differentiation, and microenvironment. The therapeutic efficacy and current applications of MSC-based kidney disease therapies are also discussed.
Collapse
|
20
|
Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother 2018; 109:2022-2034. [PMID: 30551458 DOI: 10.1016/j.biopha.2018.11.068] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/17/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023] Open
Abstract
Ischemic diseases refer to a wide range of diseases caused by reduced blood flow and a subsequently deficient oxygen and nutrient supply. The pathogenesis of ischemia is multifaceted and primarily involves inflammation, oxidative stress and an apoptotic response. Over the last decade, mesenchymal stem cells (MSCs) have been widely studied as potential cell therapy agents for ischemic diseases due to their multiple favourable functions. However, the low homing and survival rates of transplanted cells have been concerns limiting for their clinical application. Recently, increasing studies have attempted to enhance the efficacy of MSCs by various strategies including genetic modification, pretreatment, combined application and biomaterial application. The purpose of this review is to summarize these creative strategies and the progress in basic and preclinical studies.
Collapse
|
21
|
Hu C, Zhao L, Peng C, Li L. Regulation of the mitochondrial reactive oxygen species: Strategies to control mesenchymal stem cell fates ex vivo and in vivo. J Cell Mol Med 2018; 22:5196-5207. [PMID: 30160351 PMCID: PMC6201215 DOI: 10.1111/jcmm.13835] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/11/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are broadly used in cell‐based regenerative medicine because of their self‐renewal and multilineage potencies in vitro and in vivo. To ensure sufficient amounts of MSCs for therapeutic purposes, cells are generally cultured in vitro for long‐term expansion or specific terminal differentiation until cell transplantation. Although physiologically up‐regulated reactive oxygen species (ROS) production is essential for maintenance of stem cell activities, abnormally high levels of ROS can harm MSCs both in vitro and in vivo. Overall, additional elucidation of the mechanisms by which physiological and pathological ROS are generated is necessary to better direct MSC fates and improve their therapeutic effects by controlling external ROS levels. In this review, we focus on the currently revealed ROS generation mechanisms and the regulatory routes for controlling their rates of proliferation, survival, senescence, apoptosis, and differentiation. A promising strategy in future regenerative medicine involves regulating ROS generation via various means to augment the therapeutic efficacy of MSCs, thus improving the prognosis of patients with terminal diseases.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lingfei Zhao
- Kidney Disease Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Conggao Peng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Xia W, Zhuang L, Hou M. Role of lincRNA‑p21 in the protective effect of macrophage inhibition factor against hypoxia/serum deprivation‑induced apoptosis in mesenchymal stem cells. Int J Mol Med 2018; 42:2175-2184. [PMID: 30015822 DOI: 10.3892/ijmm.2018.3767] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/28/2018] [Indexed: 01/09/2023] Open
Abstract
Stem cell transplantation is a promising clinical strategy for curing ischemic cardiomyopathy. However, its efficacy is impaired by low cell survival following transplantation, partly caused by insufficient resistance of the transplanted stem cells to severe oxidative stress at the injury site. In the current study, it was demonstrated that the small‑molecule macrophage migration inhibitory factor (MIF) enhanced the defense of bone marrow‑derived mesenchymal stem cells (MSCs) against hypoxia/serum deprivation (SD)‑induced apoptosis in vitro. MIF significantly suppressed apoptosis and caspase family activities through inhibition of long intergenic noncoding (linc) RNA‑p21 to maintain activation of the Wnt/β‑catenin signaling pathway. The regulatory loop between MIF and the lincRNA‑p21‑Wnt/β‑catenin signaling pathway was identified to be associated with the inhibition of oxidative stress. The involvement of the lincRNA‑p21‑Wnt/β‑catenin signaling pathway in the effects of MIF in MSCs by overexpression of lincRNA‑p21and silencing β‑catenin using small interfering RNA was also demonstrated, both of which abolished the anti‑apoptotic and anti‑oxidative effects of MIF in MSCs under hypoxia/SD conditions. In conclusion, MIF protected MSCs from hypoxia/SD‑induced apoptosis by interacting with lincRNA‑p21, leading to activation of the downstream Wnt/β‑catenin signaling pathway and decreased oxidative stress. Thus, treatment with MIF may have important therapeutic implications in improving MSC survival and therapeutic efficiency.
Collapse
Affiliation(s)
- Wenzheng Xia
- Department of Neurosurgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Lei Zhuang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Meng Hou
- Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
23
|
Ge Z, Diao H, Ji X, Liu Q, Zhang X, Wu Q. Gap junctional intercellular communication and endoplasmic reticulum stress regulate chronic cadmium exposure induced apoptosis in HK-2 cells. Toxicol Lett 2018; 288:35-43. [DOI: 10.1016/j.toxlet.2018.02.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/23/2018] [Accepted: 02/08/2018] [Indexed: 12/23/2022]
|
24
|
Fucoidan Rescues p-Cresol-Induced Cellular Senescence in Mesenchymal Stem Cells via FAK-Akt-TWIST Axis. Mar Drugs 2018; 16:md16040121. [PMID: 29642406 PMCID: PMC5923408 DOI: 10.3390/md16040121] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/19/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a source for cell-based therapy. Although MSCs have the potential for tissue regeneration, their therapeutic efficacy is restricted by the uremic toxin, p-cresol, in chronic kidney disease (CKD). To address this issue, we investigated the effect of fucoidan, a marine sulfated polysaccharide, on cellular senescence in MSCs. After p-cresol exposure, MSC senescence was induced, as indicated by an increase in cell size and a decrease in proliferation capacity. Treatment of senescent MSCs with fucoidan significantly reversed this cellular senescence via regulation of SMP30 and p21, and increased proliferation through the regulation of cell cycle-associated proteins (CDK2, CDK4, cyclin D1, and cyclin E). These effects were dependent on FAK-Akt-TWIST signal transduction. In particular, fucoidan promoted the expression of cellular prion protein (PrPC), which resulted in the maintenance of cell expansion capacity in p-cresol-induced senescent MSCs. This protective effect of fucoidan on senescence-mediated inhibition of proliferation was dependent on the TWIST-PrPC axis. In summary, this study shows that fucoidan protects against p-cresol-induced cellular senescence in MSCs through activation of the FAK-Akt-TWIST pathway and suggests that fucoidan could be used in conjunction with functional MSC-based therapies in the treatment of CKD.
Collapse
|
25
|
Tauroursodeoxycholic Acid Protects against the Effects of P-Cresol-Induced Reactive Oxygen Species via the Expression of Cellular Prion Protein. Int J Mol Sci 2018; 19:ijms19020352. [PMID: 29370069 PMCID: PMC5855574 DOI: 10.3390/ijms19020352] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/16/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) could be a promising solution in the treatment of various diseases including chronic kidney disease (CKD). However, endoplasmic reticulum (ER) stress induced by ischemia in the area of application limits the integration and survival of MSCs in patients. In our study, we generated ER stress-induced conditions in MSCs using P-cresol. As P-cresol is a toxic compound accumulated in the body of CKD patients and induces apoptosis and inflammation through reactive oxygen species (ROS), we observed ER stress-induced MSC apoptosis activated by oxidative stress, which in turn resulted from ROS generation. To overcome stress-induced apoptosis, we investigated the protective effects of tauroursodeoxycholic acid (TUDCA), a bile acid, on ER stress in MSCs. In ER stress, TUDCA treatment of MSCs reduced ER stress-associated protein activation, including GRP78, PERK, eIF2α, ATF4, IRE1α, and CHOP. Next, to explore the protective mechanism adopted by TUDCA, TUDCA-mediated cellular prion protein (PrPC) activation was assessed. We confirmed that PrPC expression significantly increased ROS, which was eliminated by superoxide dismutase and catalase in MSCs. These findings suggest that TUDCA protects from inflammation and apoptosis in ER stress via PrPC expression. Our study demonstrates that TUDCA protects MSCs against inflammation and apoptosis in ER stress by PrPC expression in response to P-cresol exposure.
Collapse
|
26
|
Wang Z, Liu T, Chen X, You H, Zhang Q, Xue J, Zheng Y, Luo D. Low molecular weight fucoidan ameliorates hindlimb ischemic injury in type 2 diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2018; 210:434-442. [PMID: 28917976 DOI: 10.1016/j.jep.2017.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/25/2017] [Accepted: 09/12/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Low molecular weight fucoidan (LMWF), extracted from Laminaria japonica Areschoug, is a traditional Chinese medicine, commonly used to alleviate edema, particularly for feet with numbness and pain. AIM OF THE STUDY Diabetic mellitus (DM) patients are at high risk of developing peripheral arterial disease (PAD). Individuals with DM and PAD co-morbidity have a much higher risk of critical limb ischemia. LMWF showed several beneficial effects, such as anti-inflammation, anti-thrombosis, and enhancing revascularization. Therefore, we hypothesized that LMWF might be beneficial to diabetes-induced PAD, and investigated the therapeutic potential of LMWF on diabetic PAD rats. MATERIALS AND METHODS Type 2 diabetic Goto-Kakizaki (GK) rats were made PAD by injection of sodium laurate into femoral artery. LMWF (20, 40 or 80mg/kg/day) or cilostazol (100mg/kg/day) were given to diabetic PAD rats for 4 weeks, respectively. The effects of LMWF on foot ulceration and claudication, plantar blood flow, collateral vessel formation, endothelium morphology, gastrocnemius injury, platelet aggregation, vessel vasodilation, and the expressions of inflammation factors, VEGF, eNOS, and nitric oxide were measured. RESULTS We found that LMWF markedly ameliorated foot ulceration and claudication, and improved the plantar perfusion by reversing hyperreactive platelet aggregation, ameliorating endothelium-dependent vasodilation and revascularization on diabetic PAD rats. In addition, upregulation of several inflammatory factors, such as ICAM-1 and IL-1β in the gastrocnemius muscles of ischemic hindlimb were suppressed by LMWF administration. And eNOS phosphorylation at Ser1177 and NO production were significantly enhanced in LMWF-treated diabetic PAD rats. CONCLUSIONS Taken together, our findings demonstrated that LMWF exhibits therapeutic effect on hindlimb ischemia in type 2 diabetic rats likely through ameliorating endothelium eNOS dysfunction and enhancing revascularization, thus, providing a potential supplementary non-invasive treatment for diabetes-induced PAD.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China
| | - Tiantian Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China
| | - Xiaoping Chen
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China
| | - Hongjie You
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China
| | - Quanbin Zhang
- Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Jingyi Xue
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China
| | - Yuanyuan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China.
| | - Dali Luo
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, District of Feng Tai, Street of Youanmenwai, #10 Xi TouTiao, Beijing 100069, PR China.
| |
Collapse
|
27
|
Shen Y, Yan Y, Lu L, Qian Y, Guan X, Zhang L, Qi Y, Gu L, Ding F. Klotho ameliorates hydrogen peroxide-induced oxidative injury in TCMK-1 cells. Int Urol Nephrol 2017; 50:787-798. [PMID: 29285593 DOI: 10.1007/s11255-017-1765-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/05/2017] [Indexed: 12/31/2022]
Abstract
PURPOSE Defects in Klotho gene expression in mice result in a vulnerability to oxidative injuries. We aimed to identify the expression of Klotho in a mouse tubular epithelial (TCMK-1) cell line, and also to investigate changes in Klotho expression induced by oxidative stress and the potential role of intra- and extracellular Klotho protein. METHODS During exposure to hydrogen peroxide (H2O2), an overexpression of the Klotho gene was induced and exogenous Klotho protein was added in TCMK-1 cells. The generation of reactive oxidative species (ROS) was examined by flow cytometry, and cell viability was assessed by Cell Counting Kit-8. Cellular apoptosis was determined by flow cytometry and Hoechst 33258 staining followed by Western blotting to evaluate the expression of Klotho, antioxidant enzymes, and apoptosis-associated proteins. RESULTS While H2O2 significantly suppressed Klotho expression, cell viability, and the expression of antioxidant enzymes in a concentration-dependent manner, cellular apoptosis was increased and p38/MAPK and JNK/MAPK were activated. Intra- and extracellular Klotho remarkably ameliorated viability inhibition, ROS generation, and cellular apoptosis induced by H2O2. Intra- and extracellular Klotho also reversed the loss of antioxidant enzymes, the elevation of cleaved Caspase-3 and Bax/Bcl-2, and the phosphorylation of JNK/MAPK and p38/MAPK. CONCLUSIONS Klotho has posed antioxidant and anti-apoptotic effects on oxidative injuries in TCMK-1 cells, which might be partially related to its inhibition of JNK/MAPK and p38/MAPK phosphorylation and subsequent elevation of antioxidant enzymes. Increasing Klotho expression has played a protective role against oxidative stress in tubular epithelial cells.
Collapse
Affiliation(s)
- Yue Shen
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Rd., Shanghai, 200011, China
| | - Yucheng Yan
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai, 200127, China.
| | - Liming Lu
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Rd., Shanghai, 200025, China
| | - Yingying Qian
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai, 200127, China
| | - Xuejing Guan
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai, 200127, China
| | - Lulu Zhang
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Rd., Shanghai, 200011, China
| | - Yuanyuan Qi
- Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, 227 South Chongqing Rd., Shanghai, 200025, China
| | - Leyi Gu
- Department of Nephrology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai, 200127, China
| | - Feng Ding
- Department of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, 639 Zhizaoju Rd., Shanghai, 200011, China
| |
Collapse
|
28
|
Tsai SH, Wang JC, Liao WI, Hsu YJ, Lin CY, Liao MT, Huang PH, Lin SJ. Fucoidan attenuates angiotensin II-induced abdominal aortic aneurysms through the inhibition of c-Jun N-terminal kinase and nuclear factor κB activation. J Vasc Surg 2017; 68:72S-81S.e1. [PMID: 29290496 DOI: 10.1016/j.jvs.2017.09.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/17/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Rupture of abdominal aortic aneurysm (AAA) is one of the leading causes of sudden death among the elderly. Most incidental AAAs are below the threshold for intervention at the time of detection; however, there is no evidence that commonly used cardiovascular drugs have clinical beneficial effects on AAA progression. Therefore, in addition to current cardiovascular risk-reducing treatments, an adjunctive medical therapy targeting the regulation of extracellular matrix metabolism is still required in the clinical setting. Fucoidan is an extract of brown seaweed and a sulfated polysaccharide. Emerging evidence suggests that fucoidan has potential cardiovascular applications. Numerous investigations of fucoidan in diseases of the cardiovascular system have mainly focused on its pleiotropic anti-inflammatory effects. Specifically, fucoidan has been shown to have matrix metalloproteinase (MMP)-reducing effects in several studies. We aimed to evaluate the beneficial effect of fucoidan on aneurysmal growth in a murine model of aortic aneurysm and further provide a rationale for using fucoidan as a medical adjunctive therapy. METHODS A murine model of angiotensin II (Ang II)-induced AAA was used to assess the therapeutic effects of fucoidan on AAA growth in vivo. The characteristics and quantification of AAAs were determined in situ. Human umbilical vein endothelial cells were used for studying the involved pathways in vitro. Western blotting was used to detect the involved signaling pathways both in vivo and in vitro. RESULTS Treatment with fucoidan significantly reduced the incidence of AAA formation. Administration of fucoidan significantly attenuated Ang II-induced aortic expansion from 1.56 ± 0.76 mm to 1.09 ± 0.30 mm. Administration of fucoidan significantly suppressed MMP-2 and MMP-9 activities and reduced the grade of elastin degradation in vivo. In vitro, we found that fucoidan could ameliorate the Ang II-induced phosphorylation of c-Jun N-terminal kinase and nuclear factor κB p65, and it further reduced MMP and reactive oxygen species production. CONCLUSIONS Fucoidan inhibits the progression of experimental AAA growth through the attenuation of proinflammatory nuclear factor κB and c-Jun N-terminal kinase activation. Fucoidan could be a potential medical adjunctive therapy for small AAAs.
Collapse
Affiliation(s)
- Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| | - Jen-Chun Wang
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Tser Liao
- Department of Pediatrics, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
29
|
Deniaud-Bouët E, Hardouin K, Potin P, Kloareg B, Hervé C. A review about brown algal cell walls and fucose-containing sulfated polysaccharides: Cell wall context, biomedical properties and key research challenges. Carbohydr Polym 2017; 175:395-408. [PMID: 28917882 DOI: 10.1016/j.carbpol.2017.07.082] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/04/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Studies on brown algal cell walls have entered a new phase with the concomitant discovery of novel polysaccharides present in cell walls and the establishment of a comprehensive generic model for cell wall architecture. Brown algal cell walls are composites of structurally complex polysaccharides. In this review we discuss the most recent progress in the structural composition of brown algal cell walls, emphasizing the significance of extraction and screening techniques, and the biological activities of the corresponding polysaccharides, with a specific focus on the fucose-containing sulfated polysaccharides. They include valuable marine molecules that exert a broad range of pharmacological properties such as antioxidant and anti-inflammatory activities, functions in the regulation of immune responses and of haemostasis, anti-infectious and anticancer actions. We identify the key remaining challenges in this research field.
Collapse
Affiliation(s)
- Estelle Deniaud-Bouët
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France; CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France.
| | - Kevin Hardouin
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France; CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France.
| | - Philippe Potin
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France; CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France.
| | - Bernard Kloareg
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France; CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France.
| | - Cécile Hervé
- Sorbonne Universités, UPMC Univ Paris 06, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France; CNRS, UMR 8227, Integrative Biology of Marine Models, Station Biologique de Roscoff, CS 90074, Roscoff, France.
| |
Collapse
|
30
|
Lee YE, Kim H, Seo C, Park T, Lee KB, Yoo SY, Hong SC, Kim JT, Lee J. Marine polysaccharides: therapeutic efficacy and biomedical applications. Arch Pharm Res 2017; 40:1006-1020. [PMID: 28918561 PMCID: PMC7090684 DOI: 10.1007/s12272-017-0958-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/10/2017] [Indexed: 12/22/2022]
Abstract
The ocean contains numerous marine organisms, including algae, animals, and plants, from which diverse marine polysaccharides with useful physicochemical and biological properties can be extracted. In particular, fucoidan, carrageenan, alginate, and chitosan have been extensively investigated in pharmaceutical and biomedical fields owing to their desirable characteristics, such as biocompatibility, biodegradability, and bioactivity. Various therapeutic efficacies of marine polysaccharides have been elucidated, including the inhibition of cancer, inflammation, and viral infection. The therapeutic activities of these polysaccharides have been demonstrated in various settings, from in vitro laboratory-scale experiments to clinical trials. In addition, marine polysaccharides have been exploited for tissue engineering, the immobilization of biomolecules, and stent coating. Their ability to detect and respond to external stimuli, such as pH, temperature, and electric fields, has enabled their use in the design of novel drug delivery systems. Thus, along with the promising characteristics of marine polysaccharides, this review will comprehensively detail their various therapeutic, biomedical, and miscellaneous applications.
Collapse
Affiliation(s)
- Young-Eun Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Hyeongmin Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Changwon Seo
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Taejun Park
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Kyung Bin Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Seung-Yup Yoo
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Seong-Chul Hong
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Jeong Tae Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea.
| |
Collapse
|
31
|
Abstract
Background. The polysaccharide fucoidan is widely investigated as an anti-cancer agent. Here, we tested the effect of fucoidan on uveal melanoma cell lines. Methods. The effect of 100 µM fucoidan was investigated on five cell lines (92.1, Mel270 OMM1, OMM2.3, OMM2.5) and of 1 µg/mL–1 mg/mL fucoidan in two cell lines (OMM1, OMM2.3). Cell proliferation and viability were investigated with a WST-1 assay, migration in a wound healing (scratch) assay. Vascular Endothelial Growth Factor (VEGF) was measured in ELISA. Angiogenesis was evaluated in co-cultures with endothelial cells. Cell toxicity was induced by hydrogen-peroxide. Protein expression (Akt, ERK1/2, Bcl-2, Bax) was investigated in Western blot. Results. Fucoidan increased proliferation in two and reduced it in one cell line. Migration was reduced in three cell lines. The effect of fucoidan on VEGF was cell type and concentration dependent. In endothelial co-culture with 92.1, fucoidan significantly increased tubular structures. Moreover, fucoidan significantly protected all tested uveal melanoma cell lines from hydrogen-peroxide induced cell death. Under oxidative stress, fucoidan did not alter the expression of Bcl-2, Bax or ERK1/2, while inducing Akt expression in 92.1 cells but not in any other cell line. Conclusion. Fucoidan did not show anti-tumorigenic effects but displayed protective and pro-angiogenic properties, rendering fucoidan unsuitable as a potential new drug for the treatment of uveal melanoma.
Collapse
|
32
|
Kornicka K, Kocherova I, Marycz K. The effects of chosen plant extracts and compounds on mesenchymal stem cells-a bridge between molecular nutrition and regenerative medicine- concise review. Phytother Res 2017; 31:947-958. [DOI: 10.1002/ptr.5812] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Katarzyna Kornicka
- Department of Experimental Biology and Electron Microscope Facility, The Faculty of Biology and Animal Science; Wroclaw University of Environmental and Life Sciences; Chełmońskiego 38c 50-630 Wrocław Poland
- Wroclaw Research Centre EIT+; 54-066 Wrocław Poland
| | - Ievgeniia Kocherova
- Department of Experimental Biology and Electron Microscope Facility, The Faculty of Biology and Animal Science; Wroclaw University of Environmental and Life Sciences; Chełmońskiego 38c 50-630 Wrocław Poland
| | - Krzysztof Marycz
- Department of Experimental Biology and Electron Microscope Facility, The Faculty of Biology and Animal Science; Wroclaw University of Environmental and Life Sciences; Chełmońskiego 38c 50-630 Wrocław Poland
- Wroclaw Research Centre EIT+; 54-066 Wrocław Poland
| |
Collapse
|
33
|
Wu M, Gu L, Gong Q, Sun J, Ma Y, Wu H, Wang Y, Guo G, Li X, Zhu H. Strategies to reduce the intracellular effects of iron oxide nanoparticle degradation. Nanomedicine (Lond) 2017; 12:555-570. [PMID: 28181458 DOI: 10.2217/nnm-2016-0328] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a significant self-renewal capacity and can differentiate into a variety of cell types. Cell labeling is crucial as it is difficult to detect cell fate after transplantation in vivo. MSCs labeled with iron oxide nanoparticles (IONPs), which can be tracked by MRI, have tremendous potential in regenerative medicine and oncological research. As a part of nanoparticle, the iron oxide core is a key aspect that can exhibit adverse or beneficial effects on MSCs labeled for tracking. Some IONPs exhibit adverse effects, such as cytotoxicity and apoptosis, while other IONPs exhibit beneficial functions that can promote both MSC proliferation and homing efficiency. This review reveals the cytotoxic mechanisms and potential functions of the iron oxide core of IONPs in cell labeling as well as strategies for minimizing the intracellular effects of IONPs.
Collapse
Affiliation(s)
- Min Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Jiayu Sun
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yiqi Ma
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Haoxing Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yu Wang
- College of Life Science, Sichuan Normal University, Chengdu 610068, China
| | - Gang Guo
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xue Li
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Yoon YM, Lee JH, Yun SP, Han YS, Yun CW, Lee HJ, Noh H, Lee SJ, Han HJ, Lee SH. Tauroursodeoxycholic acid reduces ER stress by regulating of Akt-dependent cellular prion protein. Sci Rep 2016; 6:39838. [PMID: 28004805 PMCID: PMC5177936 DOI: 10.1038/srep39838] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/28/2016] [Indexed: 11/09/2022] Open
Abstract
Although mesenchymal stem cells (MSCs) are a promising cell source for regenerative medicine, ischemia-induced endoplasmic reticulum (ER) stress induces low MSC engraftment and limits their therapeutic efficacy. To overcome this, we investigated the protective effect of tauroursodeoxycholic acid (TUDCA), a bile acid, on ER stress in MSCs in vitro and in vivo. In ER stress conditions, TUDCA treatment of MSCs reduced the activation of ER stress-associated proteins, including GRP78, PERK, eIF2α, ATF4, IRE1α, JNK, p38, and CHOP. In particular, TUDCA inhibited the dissociation between GRP78 and PERK, resulting in reduced ER stress-mediated cell death. Next, to explore the ER stress protective mechanism induced by TUDCA treatment, TUDCA-mediated cellular prion protein (PrPC) activation was assessed. TUDCA treatment increased PrPC expression, which was regulated by Akt phosphorylation. Manganese-dependent superoxide dismutase (MnSOD) expression also increased significantly in response to signaling through the TUDCA-Akt axis. In a murine hindlimb ischemia model, TUDCA-treated MSC transplantation augmented the blood perfusion ratio, vessel formation, and transplanted cell survival more than untreated MSC transplantation did. Augmented functional recovery following MSC transplantation was blocked by PrPC downregulation. This study is the first to demonstrate that TUDCA protects MSCs against ER stress via Akt-dependent PrPC and Akt-MnSOD pathway.
Collapse
Affiliation(s)
- Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Birmingham, AL 35294, USA
| | - Seung Pil Yun
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Republic of Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan, 330-930, Republic of Korea
| |
Collapse
|
35
|
Lee SS, Lee SJ, Lee SH, Ryu JM, Lim HS, Kim JS, Song EJ, Jung YH, Lee HJ, Kim CH, Han HJ. Netrin-1-Induced Stem Cell Bioactivity Contributes to the Regeneration of Injured Tissues via the Lipid Raft-Dependent Integrin α6β4 Signaling Pathway. Sci Rep 2016; 6:37526. [PMID: 27881869 PMCID: PMC5121594 DOI: 10.1038/srep37526] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/31/2016] [Indexed: 11/17/2022] Open
Abstract
Netrin-1 (Ntn-1) is a multifunctional neuronal signaling molecule; however, its physiological significance, which improves the tissue-regeneration capacity of stem cells, has not been characterized. In the present study, we investigate the mechanism by which Ntn-1 promotes the proliferation of hUCB-MSCs with regard to the regeneration of injured tissues. We found that Ntn-1 induces the proliferation of hUCB-MSCs mainly via Inα6β4 coupled with c-Src. Ntn-1 induced the recruitment of NADPH oxidases and Rac1 into membrane lipid rafts to facilitate ROS production. The Inα6β4 signaling of Ntn-1 through ROS production is uniquely mediated by the activation of SP1 for cell cycle progression and the transcriptional occupancy of SP1 on the VEGF promoter. Moreover, Ntn-1 has the ability to induce the F-actin reorganization of hUCB-MSCs via the Inα6β4 signaling pathway. In an in vivo model, transplantation of hUCB-MSCs pre-treated with Ntn-1 enhanced the skin wound healing process, where relatively more angiogenesis was detected. The potential effect of Ntn-1 on angiogenesis is further verified by the mouse hindlimb ischemia model, where the pre-activation of hUCB-MSCs with Ntn-1 significantly improved vascular regeneration. These results demonstrate that Ntn-1 plays an important role in the tissue regeneration process of hUCB-MSC via the lipid raft-mediated Inα6β4 signaling pathway.
Collapse
Affiliation(s)
- Soo Sang Lee
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea.,SKY plastic surgery clinic, 4F, 826-23, Yeoksam-dong, Gangnam-gu, Seoul, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Hyeon Su Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Chung Hun Kim
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
36
|
Lee JH, Ryu JM, Han YS, Zia MF, Kwon HY, Noh H, Han HJ, Lee SH. Fucoidan improves bioactivity and vasculogenic potential of mesenchymal stem cells in murine hind limb ischemia associated with chronic kidney disease. J Mol Cell Cardiol 2016; 97:169-79. [PMID: 27216370 DOI: 10.1016/j.yjmcc.2016.05.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 04/28/2016] [Accepted: 05/18/2016] [Indexed: 12/13/2022]
Abstract
Chronic kidney disease (CKD) is a significant risk factor for cardiovascular and peripheral vascular disease. Although mesenchymal stem cell (MSC)-based therapy is a promising strategy for treatment of ischemic diseases associated with CKD, the associated pathophysiological conditions lead to low survival and proliferation of transplanted MSCs. To address these limitations, we investigated the effects of fucoidan, a sulfated polysaccharide, on the bioactivity of adipose tissue-derived MSCs and the potential of fucoidan-treated MSCs to improve neovascularization in ischemic tissues of CKD mice. Treatment of MSCs with fucoidan increased their proliferative potential and the expression of cell cycle-associated proteins, such as cyclin E, cyclin dependent kinase (CDK) 2, cyclin D1, and CDK4, via focal adhesion kinase and the phosphatidylinositol-4,5-bisphosphate 3-kinase-Akt axis. Moreover, fucoidan enhanced the immunomodulatory activity of MSCs through the ERK-IDO-1 signal cascade. Fucoidan was found to augment the proliferation, incorporation, and endothelial differentiation of transplanted MSCs at ischemic sites in CKD mice hind limbs. In addition, transplantation of fucoidan-treated MSCs enhanced the ratio of blood flow and limb salvage in CKD mice with hind limb ischemia. To our knowledge, our findings are the first to reveal that fucoidan enhances the bioactivity of MSCs and improves their neovascularization in ischemic injured tissues of CKD. In conclusion, fucoidan-treated MSCs may provide an important pathway toward therapeutic neovascularization in patients with CKD.
Collapse
Affiliation(s)
- Jun Hee Lee
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 626-870, Republic of Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea; Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea; Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 330-930, Republic of Korea
| | - Mohammad Farid Zia
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Hyog Young Kwon
- Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 330-930, Republic of Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Soonchunhyang University, Seoul, Republic of Korea; Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Republic of Korea.
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine and Research Institute for Veterinary Science, and BK21 PLUS Creative Veterinary Research Center, Seoul National University, Seoul 151-741, Republic of Korea.
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea; Department of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 330-930, Republic of Korea.
| |
Collapse
|
37
|
Lee JH, Han YS, Lee SH. Long-Duration Three-Dimensional Spheroid Culture Promotes Angiogenic Activities of Adipose-Derived Mesenchymal Stem Cells. Biomol Ther (Seoul) 2016; 24:260-7. [PMID: 26869524 PMCID: PMC4859789 DOI: 10.4062/biomolther.2015.146] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/05/2015] [Accepted: 11/11/2015] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) offer significant therapeutic promise for various regenerative therapies. However, MSC-based therapy for injury exhibits low efficacy due to the pathological environment in target tissues and the differences between in vitro and in vivo conditions. To address this issue, we developed adipose-derived MSC spheroids as a novel delivery method to preserve the stem cell microenvironment. MSC spheroids were generated by suspension culture for 3 days, and their sizes increased in a time-dependent manner. After re-attachment of MSC spheroids to the plastic dish, their adhesion capacity and morphology were not altered. MSC spheroids showed enhanced production of hypoxia-induced angiogenic cytokines such as vascular endothelial growth factor (VEGF), stromal cell derived factor (SDF), and hepatocyte growth factor (HGF). In addition, spheroid culture promoted the preservation of extracellular matrix (ECM) components, such as laminin and fibronectin, in a culture time- and spheroid size-dependent manner. Furthermore, phosphorylation of AKT, a cell survival signal, was significantly higher and the expression of pro-apoptotic molecules, poly (ADP ribose) polymerase-1 (PARP-1) and cleaved caspase-3, was markedly lower in the spheroids than in MSCs in monolayers. In the murine hindlimb ischemia model, transplanted MSC spheroids showed better proliferation than MSCs in monolayer. These findings suggest that MSC spheroids promote MSC bioactivities via secretion of angiogenic cytokines, preservation of ECM components, and regulation of apoptotic signals. Therefore, MSC spheroid-based cell therapy may serve as a simple and effective strategy for regenerative medicine.
Collapse
Affiliation(s)
- Jun Hee Lee
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 31151, Republic of Korea
| |
Collapse
|
38
|
Danhong Promotes Angiogenesis in Diabetic Mice after Critical Limb Ischemia by Activation of CSE-H 2 S-VEGF Axis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:276263. [PMID: 26491459 PMCID: PMC4605378 DOI: 10.1155/2015/276263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 08/21/2015] [Accepted: 09/06/2015] [Indexed: 01/18/2023]
Abstract
The aim of this paper is to investigate effect and mechanism of Danhong injection (DH) on angiogenesis in the diabetic hind limb ischemia mouse model. Thirty diabetic hind limb ischemic model mice and ten normal mice, established by intraperitoneal (i.p.) injection of streptozotocin (STZ) or PBS and ligation/excision of femoral artery, and then twenty diabetic hind limb ischemic model mice of all were evenly randomized to saline (control, n = 10) and DH i.p. injection (2 mL/kg weight for 7 days, n = 10) groups. Limb perfusion recovery and femoral blood hydrogen sulfide (H2S) and vessel regeneration and lower limb vascular endothelial growth factor (VEGF)/cystathionine γ-lyase (CSE) expression were evaluated during intervention and after euthanasia, respectively. DH i.p. increased ischemic limb perfusion and promoted collateral circulation generation without decreasing blood glucose level. Increased local CSE-H2S-VEGF expression contributed to beneficial effects of DH injection. In conclusion, activation of local CSE-H2S-VEGF axis might participate in proangiogenesis effects of DH injection in diabetic hind limb ischemia model mice, suggesting a potential therapy for diabetic patients with critical limb ischemia.
Collapse
|
39
|
Fitton JH, Stringer DN, Karpiniec SS. Therapies from Fucoidan: An Update. Mar Drugs 2015; 13:5920-46. [PMID: 26389927 PMCID: PMC4584361 DOI: 10.3390/md13095920] [Citation(s) in RCA: 235] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/02/2015] [Accepted: 09/06/2015] [Indexed: 12/30/2022] Open
Abstract
Fucoidans are a class of sulfated fucose-rich polysaccharides found in brown marine algae and echinoderms. Fucoidans have an attractive array of bioactivities and potential applications including immune modulation, cancer inhibition, and pathogen inhibition. Research into fucoidan has continued to gain pace over the last few years and point towards potential therapeutic or adjunct roles. The source, extraction, characterization and detection of fucoidan is discussed.
Collapse
Affiliation(s)
- Janet Helen Fitton
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Damien N Stringer
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| | - Samuel S Karpiniec
- Marinova Pty Ltd., 249 Kennedy Drive, Cambridge, Tasmania 7170, Australia.
| |
Collapse
|