1
|
Yao J, Liu Y, Lin H, Shao C, Jin X, Peng T, Liu Y. Caffeic acid activates Nrf2 enzymes, providing protection against oxidative damage induced by ionizing radiation. Brain Res Bull 2025; 224:111325. [PMID: 40174789 DOI: 10.1016/j.brainresbull.2025.111325] [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: 01/13/2025] [Revised: 03/15/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
Caffeic acid (CA) is a prevalent polyphenolic compound commonly found in various plant-derived foods. Due to its diverse pharmacological properties, including antioxidant activity, cardiovascular protection, and immune regulation, CA has garnered significant attention. Ionizing radiation (IR) is extensively utilized across industrial sectors, agriculture, defense applications, scientific research, and clinical medicine; however, the detrimental effects of radiation on human health cannot be ignored. IR can directly damage the DNA, proteins, and lipids within macromolecules or ionize water molecules to generate substantial quantities of free radicals that indirectly harm cells, especially those in the brain which are highly susceptible to radiation exposure. Consequently, effective strategies for preventing and treating IR-induced neurological damage represent an urgent medical challenge that necessitates resolution. Our study aims to investigate the protective effects of CA against IR-induced neuronal cell damage along with elucidating its potential mechanisms of action. The results indicate that CA can covalently modify active cysteine residues on Keap1 protein altering its conformation; this modification disrupts the interaction between Keap1 and Nrf2 while facilitating Nrf2's translocation into the nucleus where it activates downstream expression of cellular protective factors such as heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase 1 (NQO1), Thioredoxin Reductase-1 (TrxR1) and other cellular protective factors to play a role in countering radiation-induced neurological damage. In conclusion, CA emerges as an effective radioprotective agent capable of exerting antiradiation effects. Our findings provide valuable insights for developing novel therapeutic agents aimed at preventing and treating IR-induced neurological impairment.
Collapse
Affiliation(s)
- Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| | - Yuanyuan Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Huanhuan Lin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Changxin Shao
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Xiaojie Jin
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Ting Peng
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yongqi Liu
- College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| |
Collapse
|
2
|
Yang L, Li S, Hou C, Wang Z, He W, Zhang W. Recent advances in mRNA-based therapeutics for neurodegenerative diseases and brain tumors. NANOSCALE 2025; 17:3537-3548. [PMID: 39750745 DOI: 10.1039/d4nr04394d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Messenger RNA (mRNA) therapy is an innovative approach that delivers specific protein-coding information. By promoting the ribosomal synthesis of target proteins within cells, it supplements functional or antigenic proteins to treat diseases. Unlike traditional gene therapy, mRNA does not need to enter the cell nucleus, reducing the risks associated with gene integration. Moreover, protein expression levels can be regulated by adjusting the dosage and degradation rates of mRNA. As a new generation gene therapy strategy, mRNA therapy represents the latest advancements and trends in the field. It offers advantages such as precision, safety, and ease of modification. It has been widely used in the prevention of COVID-19. Unlike acute conditions such as cerebral hemorrhage and stroke that often require immediate surgical or interventional treatments, neurodegenerative diseases (NDs) and brain tumors progress relatively slowly and face challenges such as the blood-brain barrier and complex pathogenesis. These characteristics make them particularly suitable for mRNA therapy. With continued research, mRNA-based therapeutics are expected to play a significant role in the prevention and treatment of NDs and brain tumors. This paper reviews the preparation and delivery of mRNA drugs and summarizes the research progress of mRNA gene therapy in treating NDs and brain tumors. It also discusses the current challenges, providing a theoretical basis and reference for future research in this field.
Collapse
Affiliation(s)
- Lizhi Yang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Shuo Li
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Chao Hou
- Department of Ultrasound, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Wen He
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wei Zhang
- Department of Ultrasound, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Jiang MQ, Yu SP, Estaba T, Choi E, Berglund K, Gu X, Wei L. Reprogramming Glioblastoma Cells into Non-Cancerous Neuronal Cells as a Novel Anti-Cancer Strategy. Cells 2024; 13:897. [PMID: 38891029 PMCID: PMC11171681 DOI: 10.3390/cells13110897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Glioblastoma Multiforme (GBM) is an aggressive brain tumor with a high mortality rate. Direct reprogramming of glial cells to different cell lineages, such as induced neural stem cells (iNSCs) and induced neurons (iNeurons), provides genetic tools to manipulate a cell's fate as a potential therapy for neurological diseases. NeuroD1 (ND1) is a master transcriptional factor for neurogenesis and it promotes neuronal differentiation. In the present study, we tested the hypothesis that the expression of ND1 in GBM cells can force them to differentiate toward post-mitotic neurons and halt GBM tumor progression. In cultured human GBM cell lines, including LN229, U87, and U373 as temozolomide (TMZ)-sensitive and T98G as TMZ-resistant cells, the neuronal lineage conversion was induced by an adeno-associated virus (AAV) package carrying ND1. Twenty-one days after AAV-ND1 transduction, ND1-expressing cells displayed neuronal markers MAP2, TUJ1, and NeuN. The ND1-induced transdifferentiation was regulated by Wnt signaling and markedly enhanced under a hypoxic condition (2% O2 vs. 21% O2). ND1-expressing GBM cultures had fewer BrdU-positive proliferating cells compared to vector control cultures. Increased cell death was visualized by TUNEL staining, and reduced migrative activity was demonstrated in the wound-healing test after ND1 reprogramming in both TMZ-sensitive and -resistant GBM cells. In a striking contrast to cancer cells, converted cells expressed the anti-tumor gene p53. In an orthotopical GBM mouse model, AAV-ND1-reprogrammed U373 cells were transplanted into the fornix of the cyclosporine-immunocompromised C57BL/6 mouse brain. Compared to control GBM cell-formed tumors, cells from ND1-reprogrammed cultures formed smaller tumors and expressed neuronal markers such as TUJ1 in the brain. Thus, reprogramming using a single-factor ND1 overcame drug resistance, converting malignant cells of heterogeneous GBM cells to normal neuron-like cells in vitro and in vivo. These novel observations warrant further research using patient-derived GBM cells and patient-derived xenograft (PDX) models as a potentially effective treatment for a deadly brain cancer and likely other astrocytoma tumors.
Collapse
Affiliation(s)
- Michael Q. Jiang
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
- Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA 30033, USA
| | - Takira Estaba
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| | - Emily Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| | - Ken Berglund
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Xiaohuan Gu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA 30033, USA
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30033, USA; (M.Q.J.); (T.E.); (E.C.); (X.G.)
| |
Collapse
|
4
|
Zheng YY, Xu H, Wang YS. Progress in direct reprogramming of dopaminergic cell replacement therapy. Neurol Sci 2024; 45:873-881. [PMID: 37945931 DOI: 10.1007/s10072-023-07175-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: 04/24/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023]
Abstract
Parkinson's disease (PD) is a gradual neurodegenerative disease. While drug therapy and surgical treatments have been the primary means of addressing PD, they do not offer a cure, and the risks associated with surgical treatment are high. Recent advances in cell reprogramming have given rise to new prospects for the treatment of Parkinson's disease (PD), with induced pluripotent stem cells (iPSCs), induced dopamine neurons (iDNs), and induced neural stem cells (iNSCs) being created. These cells can potentially be used in the treatment of Parkinson's disease. On the other hand, this article emphasizes the limits of iPSCs and iNSCs in the context of Parkinson's disease treatment, as well as approaches for direct reprogramming of somatic cells into iDNs. The paper will examine the benefits and drawbacks of directly converting somatic cells into iDNs.
Collapse
Affiliation(s)
- Yuan Yuan Zheng
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Hui Xu
- Human Resources Department, Binzhou Medical University, Yantai, 264003, Shandong, China
| | - Yue Si Wang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, Shandong, China.
- Medicine and Pharmacy Research Center, Binzhou Medical University, Yantai, 264003, Shandong, China.
- Yantai Key Laboratory of Stem Cell and Regenerative Medicine, Binzhou Medical University, Yantai, 264003, Shandong, China.
| |
Collapse
|
5
|
Wang X, Wu J, Wang W, Zhang Y, He D, Xiao B, Zhang H, Song A, Xing Y, Li B. Reprogramming of Rat Fibroblasts into Induced Neurons by Small-Molecule Compounds In Vitro and In Vivo. ACS Chem Neurosci 2022; 13:2099-2109. [PMID: 35723446 DOI: 10.1021/acschemneuro.2c00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cell replacement is a promising approach for neurodegenerative disease treatment. Somatic cells such as fibroblasts can be induced to differentiate into neurons by specific transcription factors; however, the potential of viral vectors used for reprogramming to integrate into the genome raises concerns about the potential clinical applications of this approach. Here, we directly reprogrammed rat embryonic skin fibroblasts into induced neurons (iNs) via six small-molecule compounds (SMs) (VPA, CHIR99021, forskolin, Y-27632, Repsox, and P7C3-A20). iNs exhibit typical neuronal morphology, and immunofluorescence showed that more than 96% of the iNs expressed the early neuronal marker class III beta-tubulin (TUJ1) and that more than 91% of iNs expressed the mature neuronal marker neuron-specific enolase (NSE) after 10 days of reprogramming. Quantitative real-time polymerase chain reaction also showed that most iNs expressed the dopaminergic neuron marker tyrosine hydroxylase, the neural marker Nur correlation factor 1, the (γ-aminobutyric acid, GABA) GABAergic neuronal marker GABA, and the cholinergic neuron marker choline acetyltransferase. In addition, we found that cell proliferation decreased during reprogramming and that protein synthesis increased initially and then decreased. SMs were mixed with hydrogels, and the hydrogels were implanted subcutaneously into the backs of rats. After 7 days, the TUJ1 and NSE proteins were expressed in surrounding tissues, indicating that SMs caused reprogramming in vivo. In summary, rat skin fibroblasts can be efficiently reprogrammed into iNs by SMs in vitro and in vivo.
Collapse
Affiliation(s)
- Xueyun Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Jing Wu
- Department of Paediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001 Henan, P.R. China
| | - Wang Wang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Yuanwang Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Dixin He
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Boying Xiao
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Haohao Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Anqi Song
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Ying Xing
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| | - Bo Li
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000 Henan, P.R. China
| |
Collapse
|
6
|
Ma K, Deng X, Xia X, Fan Z, Qi X, Wang Y, Li Y, Ma Y, Chen Q, Peng H, Ding J, Li C, Huang Y, Tian C, Zheng JC. Direct conversion of mouse astrocytes into neural progenitor cells and specific lineages of neurons. Transl Neurodegener 2018; 7:29. [PMID: 30410751 PMCID: PMC6217767 DOI: 10.1186/s40035-018-0132-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/11/2018] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Cell replacement therapy has been envisioned as a promising treatment for neurodegenerative diseases. Due to the ethical concerns of ESCs-derived neural progenitor cells (NPCs) and tumorigenic potential of iPSCs, reprogramming of somatic cells directly into multipotent NPCs has emerged as a preferred approach for cell transplantation. METHODS Mouse astrocytes were reprogrammed into NPCs by the overexpression of transcription factors (TFs) Foxg1, Sox2, and Brn2. The generation of subtypes of neurons was directed by the force expression of cell-type specific TFs Lhx8 or Foxa2/Lmx1a. RESULTS Astrocyte-derived induced NPCs (AiNPCs) share high similarities, including the expression of NPC-specific genes, DNA methylation patterns, the ability to proliferate and differentiate, with the wild type NPCs. The AiNPCs are committed to the forebrain identity and predominantly differentiated into glutamatergic and GABAergic neuronal subtypes. Interestingly, additional overexpression of TFs Lhx8 and Foxa2/Lmx1a in AiNPCs promoted cholinergic and dopaminergic neuronal differentiation, respectively. CONCLUSIONS Our studies suggest that astrocytes can be converted into AiNPCs and lineage-committed AiNPCs can acquire differentiation potential of other lineages through forced expression of specific TFs. Understanding the impact of the TF sets on the reprogramming and differentiation into specific lineages of neurons will provide valuable strategies for astrocyte-based cell therapy in neurodegenerative diseases.
Collapse
Affiliation(s)
- Kangmu Ma
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Xiaobei Deng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Zhaohuan Fan
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Xinrui Qi
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yongxiang Wang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yuju Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Yizhao Ma
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Qiang Chen
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Hui Peng
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jianqing Ding
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Chunhong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Changhai Tian
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| | - Jialin C. Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People’s Hospital affiliated to Tongji University School of Medicine, Shanghai, 200072 China
- Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092 China
- Departments of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-5930 USA
| |
Collapse
|
7
|
Zhang B, Yan W, Zhu Y, Yang W, Le W, Chen B, Zhu R, Cheng L. Nanomaterials in Neural-Stem-Cell-Mediated Regenerative Medicine: Imaging and Treatment of Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1705694. [PMID: 29543350 DOI: 10.1002/adma.201705694] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/17/2017] [Indexed: 05/24/2023]
Abstract
Patients are increasingly being diagnosed with neuropathic diseases, but are rarely cured because of the loss of neurons in damaged tissues. This situation creates an urgent clinical need to develop alternative treatment strategies for effective repair and regeneration of injured or diseased tissues. Neural stem cells (NSCs), highly pluripotent cells with the ability of self-renewal and potential for multidirectional differentiation, provide a promising solution to meet this demand. However, some serious challenges remaining to be addressed are the regulation of implanted NSCs, tracking their fate, monitoring their interaction with and responsiveness to the tissue environment, and evaluating their treatment efficacy. Nanomaterials have been envisioned as innovative components to further empower the field of NSC-based regenerative medicine, because their unique physicochemical characteristics provide unparalleled solutions to the imaging and treatment of diseases. By building on the advantages of nanomaterials, tremendous efforts have been devoted to facilitate research into the clinical translation of NSC-based therapy. Here, recent work on emerging nanomaterials is highlighted and their performance in the imaging and treatment of neurological diseases is evaluated, comparing the strengths and weaknesses of various imaging modalities currently used. The underlying mechanisms of therapeutic efficacy are discussed, and future research directions are suggested.
Collapse
Affiliation(s)
- Bingbo Zhang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Wei Yan
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Key Laboratory of Green Preparation and Application for Functional Materials, Ministry of Education, School of Materials Science & Engineering, Hubei University, Wuhan, 430062, China
| | - Yanjing Zhu
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Weitao Yang
- Institute of Photomedicine, Shanghai Skin Disease Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Wenjun Le
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, 200443, China
| | - Rongrong Zhu
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| | - Liming Cheng
- Department of Spine Surgery, Tongji Hospital, Institute of Spine and Spinal Cord Injury, Tongji University School of Medicine, Tongji University, Shanghai, 200065, China
| |
Collapse
|
8
|
Strange K. Drug Discovery in Fish, Flies, and Worms. ILAR J 2017; 57:133-143. [PMID: 28053067 DOI: 10.1093/ilar/ilw034] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 10/21/2016] [Indexed: 12/22/2022] Open
Abstract
Nonmammalian model organisms such as the nematode Caenorhabditis elegans, the fruit fly Drosophila melanogaster, and the zebrafish Danio rerio provide numerous experimental advantages for drug discovery including genetic and molecular tractability, amenability to high-throughput screening methods and reduced experimental costs and increased experimental throughput compared to traditional mammalian models. An interdisciplinary approach that strategically combines the study of nonmammalian and mammalian animal models with diverse experimental tools has and will continue to provide deep molecular and genetic understanding of human disease and will significantly enhance the discovery and application of new therapies to treat those diseases. This review will provide an overview of C. elegans, Drosophila, and zebrafish biology and husbandry and will discuss how these models are being used for phenotype-based drug screening and for identification of drug targets and mechanisms of action. The review will also describe how these and other nonmammalian model organisms are uniquely suited for the discovery of drug-based regenerative medicine therapies.
Collapse
Affiliation(s)
- Kevin Strange
- Kevin Strange, Ph.D., is President and CEO of the MDI Biological Laboratory and CEO of Novo Biosciences, Inc
| |
Collapse
|
9
|
Effects of Neuropeptide Y on Stem Cells and Their Potential Applications in Disease Therapy. Stem Cells Int 2017; 2017:6823917. [PMID: 29109742 PMCID: PMC5646323 DOI: 10.1155/2017/6823917] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/01/2017] [Accepted: 08/08/2017] [Indexed: 01/04/2023] Open
Abstract
Neuropeptide Y (NPY), a 36-amino acid peptide, is widely distributed in the central and peripheral nervous systems and other peripheral tissues. It takes part in regulating various biological processes including food intake, circadian rhythm, energy metabolism, and neuroendocrine secretion. Increasing evidence indicates that NPY exerts multiple regulatory effects on stem cells. As a kind of primitive and undifferentiated cells, stem cells have the therapeutic potential to replace damaged cells, secret paracrine molecules, promote angiogenesis, and modulate immunity. Stem cell-based therapy has been demonstrated effective and considered as one of the most promising treatments for specific diseases. However, several limitations still hamper its application, such as poor survival and low differentiation and integration rates of transplanted stem cells. The regulatory effects of NPY on stem cell survival, proliferation, and differentiation may be helpful to overcome these limitations and facilitate the application of stem cell-based therapy. In this review, we summarized the regulatory effects of NPY on stem cells and discussed their potential applications in disease therapy.
Collapse
|