1
|
Lee JY, Kim ES, Kim SY, Cho YJ, Jo KH, Han JH, Moon SD. The nature and pathological impact of the c.1748A > G variant of the neurofibromin 1 gene. Gene 2025; 952:149381. [PMID: 40037421 DOI: 10.1016/j.gene.2025.149381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/17/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder, and mutations in the NF1 gene lead to RAS overactivation, which stimulates abnormal cell proliferation and can cause various tumors. The c.1748A > G mutation in the NF1 gene was initially classified as a missense mutation, but has also been suggested to be a splice mutation. It is thought that the substitution of A for G generates a cryptic splice site, resulting in a 27 bp deletion in the mRNA transcript, but this conclusion has not been documented in currently available databases. The present study was conducted to establish whether the NF1 c.1748A > G mutation induces a splicing error, and to determine whether it is pathogenic i.e. activates RAS and increases the expression of NF1-related downstream signaling molecules. We have confirmed by RT-PCR analysis of NF1 transcripts produced in the patient's peripheral blood lymphocytes as well as in a minigene construct and in iPSCs harboring the c.1748A > G mutation that this mutation creates a cryptic splice site which has the effect of deleting the first 27 bases of exon 16, and leading to transcriptional haploinsufficiency. Additionally, NPCs expressing the splicing mutant exhibited increased phosphorylation of NF1-related AKT/mTOR and Raf/MEK/Erk, as well as more effective wound healing and chemotaxis. We conclude that the NF1 c.1748A > G mutation acts as a splice mutation forming a novel cryptic site, causing a 27 bp deletion in the mRNA. This leads to increased expression of NF1-related downstream signaling molecules through RAS activation, inducing cell proliferation and potential tumor formation.
Collapse
Affiliation(s)
- Ji-Young Lee
- Division of Endocrinology and Metabolism, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, South Korea
| | - Eun Sook Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 56, Dongsu-ro, Bupyeong-gu, Incheon 21431, South Korea
| | - Su Yeon Kim
- Division of Endocrinology and Metabolism, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, South Korea
| | - Yun-Jung Cho
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 56, Dongsu-ro, Bupyeong-gu, Incheon 21431, South Korea
| | - Kwan Hoon Jo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 56, Dongsu-ro, Bupyeong-gu, Incheon 21431, South Korea
| | - Je Ho Han
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 56, Dongsu-ro, Bupyeong-gu, Incheon 21431, South Korea
| | - Sung-Dae Moon
- Division of Endocrinology and Metabolism, Institute of Biomedical Industry, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, South Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 56, Dongsu-ro, Bupyeong-gu, Incheon 21431, South Korea.
| |
Collapse
|
2
|
Mallya D, Gadre MA, Varadharajan S, Vasanthan KS. 3D bioprinting for the construction of drug testing models-development strategies and regulatory concerns. Front Bioeng Biotechnol 2025; 13:1457872. [PMID: 40028291 PMCID: PMC11868281 DOI: 10.3389/fbioe.2025.1457872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025] Open
Abstract
A drug to be successfully launched in the market requires a significant amount of capital, resources and time, where the unsuccessful results in the last stages lead to catastrophic failure for discovering drugs. This is the very reason which calls for the invention of innovative models that can closely mimic the human in vivo model for producing reliable results. Throughout the innovation line, there has been improvement in the rationale in silico designing but yet there is requirement for in vitro-in vivo correlations. During the evolving of the drug testing models, the 3D models produced by different methods have been proven to produce better results than the traditional 2D models. However, the in vitro fabrications of live tissues are still bottleneck in realizing their complete potential. There is an urgent need for the development of single, standard and simplified in vitro 3D tissue models that can be reliable for investigating the biological and pathological aspects of drug discovery, which is yet to be achieved. The existing pre-clinical models have considerable drawbacks despite being the gold standard in pre-clinical research. The major drawback being the interspecies differences and low reliability on the generated results. This gap could be overcome by the fabrication of bioengineered human disease models for drug screening. The advancement in the fabrication of 3D models will provide a valuable tool in screening drugs at different stages as they are one step closer to bio-mimic human tissues. In this review, we have discussed on the evolution of preclinical studies, and different models, including mini tissues, spheroids, organoids, bioengineered three dimensional models and organs on chips. Furthermore, we provide details of different disease models fabricated across various organs and their applications. In addition to this, the review also focuses on the limitations and the current prospects of the role of three dimensionally bioprinted models in drug screening and development.
Collapse
Affiliation(s)
- Divya Mallya
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Mrunmayi Ashish Gadre
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - S. Varadharajan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
3
|
Wang F, Liu J. The dual anti-inflammatory and anticoagulant effects of Jianpi Huashi Tongluo prescription on Rheumatoid Arthritis through inhibiting the activation of the PI3K/AKT signaling pathway. Front Pharmacol 2025; 16:1541314. [PMID: 40012623 PMCID: PMC11860884 DOI: 10.3389/fphar.2025.1541314] [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: 12/20/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is often accompanied by abnormal changes in inflammatory responses and coagulation-fibrinolysis indicators. Jianpi Huashi Tongluo Prescription - Xinfeng Capsule (XFC), a traditional Chinese medicine formulation comprising multiple herbal ingredients, is widely used clinically for the treatment of RA. It exhibits dual anti-inflammatory and anticoagulant effects. However, the specific mechanisms underlying its actions remain to be further investigated. Objective This study aims to elucidate the anti-inflammatory and anticoagulant mechanisms of XFC in the treatment of RA. Methods A multidimensional methodological framework was employed. Firstly, through retrospective clinical data mining, combined with the Apriori algorithm and random walk models, an in-depth analysis was conducted to explore the potential associations between XFC treatment and improvements in clinical inflammatory and coagulation markers among RA patients. Secondly, an adjuvant-induced arthritis rat model was established to directly observe the anti-inflammatory and anticoagulant effects of XFC in vivo. Furthermore, bioinformatics and network pharmacology techniques were applied to decipher the major active components and their targets of XFC. Lastly, a co-culture system of RA patient-derived peripheral blood mononuclear cells (RA-PBMCs) and vascular endothelial cells (VECs) was established to mimic the in vivo microenvironment, and the anti-inflammatory and anticoagulant mechanisms of XFC were validated in vitro. Results Data mining analysis revealed abnormally elevated levels of inflammatory and coagulation markers such as fibrinogen (FBG), erythrocyte sedimentation rate (ESR), high-sensitivity C-reactive protein (Hs-CRP), and rheumatoid factor (RF) in RA patients (p < 0.001), and emphasized the close correlation between XFC treatment and the improvement of these markers including Hs-CRP, ESR, and RF (confidence >60% and lift >1). Animal experimental data indicated that XFC effectively reduced the levels of inflammatory and coagulant markers (IL-6, D-D, FBG, PAF, VEGF, and TF) in adjuvant-induced arthritis (AA) rats while enhancing the expression of anti-inflammatory factors (IL-10) (p < 0.05). Furthermore, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) results suggested that the pharmacodynamic mechanism of XFC may be closely related to the regulation of the PI3K/AKT signaling pathway. Additionally, network pharmacology and molecular docking results show that the main active components of XFC, namely, calycosin-7-O-beta-D-glucoside, calycosin, and formononetin, exhibit excellent docking with the core targets HIF1A, PTGS2, and MMP9. In vitro co-culture model showed that XFC inhibited RA-related inflammatory responses and hypercoagulable states by suppressing the activation of the PI3K/AKT signaling pathway. Conclusion This study demonstrates that XFC exerts its dual anti-inflammatory and anticoagulant effects, at least in part, by inhibiting the activation of the PI3K/AKT signaling pathway, providing potential insights into targeted therapy for RA.
Collapse
Affiliation(s)
- Fanfan Wang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, First Clinical Medical College, Hefei, Anhui, China
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jian Liu
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
4
|
Liu W, Jiang H, Chen J, Tian Y, He Y, Jiao Y, Guan Y, Jia Z, Wu Y, Huang C, Ouyang Y, Xu W, Qi J, Peng J, Wang A. High paracrine activity of hADSCs cartilage microtissues inhibits extracellular matrix degradation and promotes cartilage regeneration. Mater Today Bio 2025; 30:101372. [PMID: 39839494 PMCID: PMC11745967 DOI: 10.1016/j.mtbio.2024.101372] [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: 09/04/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 01/23/2025] Open
Abstract
Due to its unique structure, articular cartilage has limited self-repair capacity. Microtissues are tiny tissue clusters that can mimic the function of target organs or tissues. Using cells alone for microtissue construction often results in the formation of necrotic cores. However, the extracellular matrix (ECM) of native cartilage can provide structural support and is an ideal source of microcarriers. Autologous adipose-derived mesenchymal stem cells (ADSCs) and bone marrow mesenchymal stem cells (BMSCs) are widely used in cartilage tissue engineering. In this study, we fabricated microcarriers and compared the behavior of two homologous cell types in the microcarrier environment. The microcarrier environment highlighted the advantages of ADSCs and promoted the proliferation and migration of these cells. Then, ADSCs microtissues (ADSCs-MT) and BMSCs microtissues (BMSCs-MT) were fabricated using a three-dimensional dynamic culture system. In vitro and in vivo experiments verified that the cartilage regeneration ability of ADSCs-MT was significantly superior to that of BMSCs-MT. Transcriptomics revealed that ADSCs-MT showed significantly lower expression levels of ECM degradation, osteogenesis, and fibrocartilage markers. Finally, the protective effect of microtissues on inflammatory chondrocytes was validated. Overall, the ADSCs-MT constructed in this study achieved excellent cartilage regeneration and could be promising for the autologous application of cartilage microtissues.
Collapse
Affiliation(s)
- Wei Liu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Hongyu Jiang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Department of Orthopedic, The Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Jiajie Chen
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Yue Tian
- The Second Medical Center of Chinese PLA General Hospital, PR China
| | - Ying He
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Ying Jiao
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Zhibo Jia
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Yanbin Wu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Cheng Huang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Department of Orthopedic, The Affiliated Hospital, Southwest Medical University, Luzhou, PR China
| | - Yiben Ouyang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Wenjing Xu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jianhong Qi
- College of Sports Medicine and Rehabilitation, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271016, PR China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| | - Aiyuan Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin, 300071, PR China
| |
Collapse
|
5
|
Panchuk I, Smirnikhina S. Toolbox for creating three-dimensional liver models. Biochem Biophys Res Commun 2024; 731:150375. [PMID: 39018971 DOI: 10.1016/j.bbrc.2024.150375] [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: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Research within the hepato-biliary system and hepatic function is currently experiencing heightened interest, this is due to the high frequency of relapse rates observed in chronic conditions, as well as the imperative for the development of innovative therapeutic strategies to address both inherited and acquired diseases within this domain. The most commonly used sources for studying hepatocytes include primary human hepatocytes, human hepatic cancer cell lines, and hepatic-like cells derived from induced pluripotent stem cells. However, a significant challenge in primary hepatic cell culture is the rapid decline in their phenotypic characteristics, dedifferentiation and short cultivation time. This limitation creates various problems, including the inability to maintain long-term cell cultures, which can lead to failed experiments in drug development and the creation of relevant disease models for researchers' purposes. To address these issues, the creation of a powerful 3D cell model could play a pivotal role as a personalized disease model and help reduce the use of animal models during certain stages of research. Such a cell model could be used for disease modelling, genome editing, and drug discovery purposes. This review provides an overview of the main methods of 3D-culturing liver cells, including a discussion of their characteristics, advantages, and disadvantages.
Collapse
Affiliation(s)
- Irina Panchuk
- Research Centre for Medical Genetics, Moscow, Russian Federation.
| | | |
Collapse
|
6
|
Zhao L, Zheng K, Wan X, Xiao Q, Yuan L, Wu C, Bao J. Chinese traditional medicine DZGP beneficially affects gut microbiome, serum metabolites and recovery from rheumatoid arthritis through mediating NF-κB signaling pathway. Heliyon 2024; 10:e33706. [PMID: 39071566 PMCID: PMC11283109 DOI: 10.1016/j.heliyon.2024.e33706] [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: 01/21/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is globally treated with several commercially available anti-inflammatory and analgesic drugs, which pose adverse side effects in many cases. Due to increasing population affected by autoimmune disorder of joints inflammation, it is crucial to use natural therapies, which are less toxic at metabolic level and promote gut health. In this study, we investigated the potential role of a locally developed traditional Chinese medicine (TCM), namely Duzheng tablet (DZGP) in controlling the RA. For this purpose, we introduced RA in male mice and divided them into 5 different groups. High throughput transcriptome analysis of synovial cells after DZGP treatment in arthritic mice revealed a significant alteration of gene expression. The correlation analysis of transcriptome with metabolites revealed that DZGP specifically targeted the B cells mediated immunity pathways. Treatment with DZGP inhibited the cytokines production, while reducing the production of inflammatory TNF-α, which led to the alleviation of inflammatory response in arthritic mice. Additionally, we applied integrated approach using 16S rDNA sequencing to understand the microbial population in relation to metabolites accumulation. The results showed that DZGP promoted the healthy gut microbiota by maintaining the ratio of Firmicutes and Bacteroidota and introduction of two additional phyla namely, Verrucomicrobiota and Cyanobacteria. Therefore, it is concluded that DZGP offers an advantage over commercial drug by changing the metabolic profile, gut microbiota while exhibiting lower cellular toxicity.
Collapse
Affiliation(s)
- Liming Zhao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei Minzu University, 445000, Enshi, China
| | - Kai Zheng
- Forest Seedlings and Wildlife Protection Management Station of Enshi Tujia and Miao Autonomous Prefecture, 445000, Enshi, China
| | - Xiaolin Wan
- College of Forestry and Horticulture, Hubei Minzu University, 445000, Enshi, China
| | - Qiang Xiao
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei Minzu University, 445000, Enshi, China
| | - Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, 445000, Enshi, China
| | - Chuanfang Wu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
| | - Jinku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, China
| |
Collapse
|
7
|
Kim D, Heo J, Song B, Lee G, Hong C, Jiang Z, Lee S, Lee K, Kim M, Park MH. 3D in vitro synovial hyperplasia model on polycaprolactone-micropatterned nanofibrous microwells for screening disease-modifying anti-rheumatic drugs. Mater Today Bio 2024; 26:101061. [PMID: 38711937 PMCID: PMC11070697 DOI: 10.1016/j.mtbio.2024.101061] [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: 12/29/2023] [Revised: 03/10/2024] [Accepted: 04/13/2024] [Indexed: 05/08/2024] Open
Abstract
Rheumatoid arthritis (RA) is known to be caused by autoimmune disorders and can be partially alleviated through Disease-Modifying Antirheumatic Drugs (DMARDs) therapy. However, due to significant variations in the physical environment and condition of each RA patient, the types and doses of DMARDs prescribed can differ greatly. Consequently, there is a need for a platform based on patient-derived cells to determine the effectiveness of specific DMARDs for individual patient. In this study, we established an RA three-dimensional (3D) spheroid that mimics the human body's 3D environment, enabling high-throughput assays by culturing patient-derived synovial cells on a macroscale-patterned polycaprolactone (PCL) scaffold. Fibroblast-like synoviocytes (FLSs) from patient and human umbilical vein endothelial cells (HUVECs) were co-cultured to simulate vascular delivery. Additionally, RA characteristics were identified at both the genetic and cytokine levels using real-time polymerase chain reaction (RT-qPCR) and dot blot assay. The similarities in junctions and adhesion were demonstrated in both actual RA patient tissues and 3D spheroids. The 3D RA spheroid was treated with representative DMARDs, observing changes in reactive oxygen species (ROS) levels, lactate dehydrogenase (LDH) levels, and inflammatory cytokine responses to confirm the varying cell reactions depending on the DMARDs used. This study underscores the significance of the 3D drug screening platform, which can be applied to diverse inflammatory disease treatments as a personalized drug screening system. We anticipate that this platform will become an indispensable tool for advancing and developing personalized DMARD treatment strategies.
Collapse
Affiliation(s)
- Dongwoo Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Jiyeon Heo
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Boa Song
- THEDONEE Inc., Research Center, Seoul, Republic of Korea
| | - Gyubok Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Changgi Hong
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Zhuomin Jiang
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Sohui Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
- Research Institute for Convergence Science, 145, Gwanggyo-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, Republic of Korea
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine, Gyeongsang National University Hospital, Jinju, 52727, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
- Department of Internal Medicine, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Min Hee Park
- THEDONEE Inc., Research Center, Seoul, Republic of Korea
| |
Collapse
|
8
|
Ren R, Jiang J, Li X, Zhang G. Research progress of autoimmune diseases based on induced pluripotent stem cells. Front Immunol 2024; 15:1349138. [PMID: 38720903 PMCID: PMC11076788 DOI: 10.3389/fimmu.2024.1349138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
Autoimmune diseases can damage specific or multiple organs and tissues, influence the quality of life, and even cause disability and death. A 'disease in a dish' can be developed based on patients-derived induced pluripotent stem cells (iPSCs) and iPSCs-derived disease-relevant cell types to provide a platform for pathogenesis research, phenotypical assays, cell therapy, and drug discovery. With rapid progress in molecular biology research methods including genome-sequencing technology, epigenetic analysis, '-omics' analysis and organoid technology, large amount of data represents an opportunity to help in gaining an in-depth understanding of pathological mechanisms and developing novel therapeutic strategies for these diseases. This paper aimed to review the iPSCs-based research on phenotype confirmation, mechanism exploration, drug discovery, and cell therapy for autoimmune diseases, especially multiple sclerosis, inflammatory bowel disease, and type 1 diabetes using iPSCs and iPSCs-derived cells.
Collapse
Affiliation(s)
| | | | | | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| |
Collapse
|
9
|
Shoda H, Natsumoto B, Fujio K. Investigation of immune-related diseases using patient-derived induced pluripotent stem cells. Inflamm Regen 2023; 43:51. [PMID: 37876023 PMCID: PMC10594759 DOI: 10.1186/s41232-023-00303-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 10/26/2023] Open
Abstract
The precise pathogenesis of immune-related diseases remains unclear, and new effective therapeutic choices are required for the induction of remission or cure in these diseases. Basic research utilizing immune-related disease patient-derived induced pluripotent stem (iPS) cells is expected to be a promising platform for elucidating the pathogenesis of the diseases and for drug discovery. Since autoinflammatory diseases are usually monogenic, genetic mutations affect the cell function and patient-derived iPS cells tend to exhibit disease-specific phenotypes. In particular, iPS cell-derived monocytic cells and macrophages can be used for functional experiments, such as inflammatory cytokine production, and are often employed in research on patients with autoinflammatory diseases.On the other hand, the utilization of disease-specific iPS cells is less successful for research on autoimmune diseases. One reason for this is that autoimmune diseases are usually polygenic, which makes it challenging to determine which factors cause the phenotypes of patient-derived iPS cells are caused by. Another reason is that protocols for differentiating some lymphocytes associated with autoimmunity, such as CD4+T cells or B cells, from iPS cells have not been well established. Nevertheless, several groups have reported studies utilizing autoimmune disease patient-derived iPS cells, including patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), and systemic sclerosis. Particularly, non-hematopoietic cells, such as fibroblasts and cardiomyocytes, differentiated from autoimmune patient-derived iPS cells have shown promising results for further research into the pathogenesis. Recently, our groups established a method for differentiating dendritic cells that produce interferon-alpha, which can be applied as an SLE pathological model. In summary, patient-derived iPS cells can provide a promising platform for pathological research and new drug discovery in the field of immune-related diseases.
Collapse
Affiliation(s)
- Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan.
| | - Bunki Natsumoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehirocho, Tsurumi-Ku, Yokohama, Kanagawa, 230-0045, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-Ku, Tokyo, 113-8655, Japan
| |
Collapse
|
10
|
Wang C, Zhao S, Xu Y, Sun W, Feng Y, Liang D, Guan Y. Integrated Microbiome and Metabolome Analysis Reveals Correlations Between Gut Microbiota Components and Metabolic Profiles in Mice with Methotrexate-Induced Hepatoxicity. Drug Des Devel Ther 2022; 16:3877-3891. [PMID: 36388083 PMCID: PMC9653027 DOI: 10.2147/dddt.s381667] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Purpose We designed this study to investigate the potential correlations between gut microbiota compositions and hepatic metabolomic disorders in mice with methotrexate (MTX)-induced hepatoxicity. Methods We used MTX to induce hepatoxicity in healthy Kunming mice, and we determined plasma ALT and AST levels and assessed the liver tissue histopathology. We applied an integrated gas chromatography-mass spectrometry (GC-MS) and 16S ribosomal RNA (rRNA) gene sequencing approach to evaluate the effects of MTX on the gut microbiota and hepatic metabolic profiles of mice. We uncovered correlations between the gut microbiota and hepatic metabolomic profiles by calculating the Spearman correlation coefficient. Results MTX caused ALT and AST level elevations and hepatoxicity in our mouse model. MTX disrupted amino acid metabolic pathways (including biosyntheses of valine, leucine, and isoleucine; and arginine; and, metabolism of alanine, aspartate, and glutamate; histidine; beta-alanine; and glycine, serine, and threonine); biosyntheses of aminoacyl-tRNA; and pantothenate, and CoA; and, metabolic pathways of energy, glutathione, and porphyrin; and chlorophyll. In addition, MTX increased the abundances of Staphylococcus, Enterococcus, Collinsella, Streptococcus, and Aerococcus, but decreased the amounts of Lactobacillus, Ruminococcus, norank_f_Muribaculaceae, unclassified_f_Lachnospiraceae, norank_f_Lachnospiraceae, A2, Eubacterium_xylanophilum_group, Phascolarctobacterium, Bifidobacterium, and Faecalibaculum. Our correlation analyses showed that different flora abundance changes including those of Phascolarctobacterium, Faecalibaculum, norank_f_Muribaculaceae, Streptococcus, Enterococcus, Staphylococcus, and Collinsella were associated with liver injury. Conclusion We present evidence supporting the notion that MTX causes hepatoxicity by altering the gut microbiota and hepatic metabolite profiles, our findings provide new venues for the management of MTX-induced hepatoxicity.
Collapse
Affiliation(s)
- Changshui Wang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272000, People’s Republic of China
| | - Shuzhen Zhao
- Children’s Rehabilitation Center, Jining Maternal and Child Health Family Planning Service Center, Jining, 272000, People’s Republic of China
| | - Yuan Xu
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Wenxue Sun
- Institute of Clinical Pharmacy and Pharmacology, Jining NO. 1 People’s Hospital, Jining Medical University, Jining, 272000, People’s Republic of China
| | - Yuanyuan Feng
- Children’s Rehabilitation Center, Jining Maternal and Child Health Family Planning Service Center, Jining, 272000, People’s Republic of China
| | - Deshuai Liang
- Department of pharmacy, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
| | - Yun Guan
- Department of Hematology, Jining NO. 1 People’s Hospital, Jining, 272000, People’s Republic of China
- Correspondence: Yun Guan; Deshuai Liang, Jining NO. 1 People’s Hospital, 6 Jiankang Road, Jining, Shandong, 272000, People’s Republic of China, Tel/Fax +86-0537 2087092, Email ;
| |
Collapse
|
11
|
Jasim SA, Yumashev AV, Abdelbasset WK, Margiana R, Markov A, Suksatan W, Pineda B, Thangavelu L, Ahmadi SH. Shining the light on clinical application of mesenchymal stem cell therapy in autoimmune diseases. Stem Cell Res Ther 2022; 13:101. [PMID: 35255979 PMCID: PMC8900359 DOI: 10.1186/s13287-022-02782-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/17/2022] [Indexed: 02/08/2023] Open
Abstract
The autoimmune diseases are associated with the host immune system, chronic inflammation, and immune reaction against self-antigens, which leads to the injury and failure of several tissues. The onset of autoimmune diseases is related to unbalanced immune homeostasis. Mesenchymal stem cells (MSCs) are multipotent cells which have capability to self-renew and differentiate into various cell types that exert a critical role in immunomodulation and regenerative therapy. Under the certain condition in vitro, MSCs are able to differentiate into multiple lineage such as osteoblasts, adipocytes, and neuron-like cells. Consequently, MSCs have a valuable application in cell treatment. Accordingly, in this review we present the last observations of researches on different MSCs and their efficiency and feasibility in the clinical treatment of several autoimmune disorders including rheumatoid arthritis, type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, inflammatory bowel disease, autoimmune liver disease, and Sjogren’s syndrome.
![]()
Collapse
Affiliation(s)
- Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-Maarif University College, Al-Anbar-Ramadi, Iraq
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia.,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation.,Industrial University, Tyumen, Russian Federation
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Benjamin Pineda
- Department of Neuroimmunology, National Institute of Neurology and Neurosurgery "Manuel Velasco Suarez" (INNN), 14269, Mexico City, Mexico
| | - Lakshmi Thangavelu
- Center for Transdisciplinary Research ,Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Seyed Hossein Ahmadi
- Cellular and Molecular Research Center, School of Medicine, Tehran University of Medical Sciences, PO Box: 1417613151, Tehran, Iran.
| |
Collapse
|
12
|
Wang J, Yuan W, Shen Q, Wu Q, Jiang Z, Wu W, Zhang L, Huang X. The key role of organic anion transporter 3 in the drug-drug interaction between tranilast and methotrexate. J Biochem Mol Toxicol 2022; 36:e22983. [PMID: 35019195 DOI: 10.1002/jbt.22983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/26/2021] [Accepted: 11/01/2021] [Indexed: 11/11/2022]
Abstract
Tranilast, N-(3',4'-dimethoxycinnamoyl)-anthranilic acid, is an anti-allergic drug and is considered for use in the treatment of rheumatoid arthritis. Methotrexate, an antimetabolite and folate antagonist to treat some cancers, is also a first-line drug for RA. The aim of this study was to understand whether tranilast could inhibit renal uptake transporters (Oat1, Oat3, and Oct2) and whether MTX combined with TL would have drug-drug interactions. The results of kidney slices and HEK293T-OAT3 cell uptake experiments showed that TL (10 μM) could inhibit the uptake of penicillin G and MTX, which are substrates of OAT3. When TL (10 mg/kg) was combined with MTX (5 mg/kg), the area under the curve and peak concentration of MTX increased by 46.46% and 113.51%, respectively, while the pharmacokinetic process of tranilast (10 mg/kg) was not changed by methotrexate (5 mg/kg). TL could increase plasma exposure of MTX by inhibiting Oat3 in vitro and in vivo.
Collapse
Affiliation(s)
- Jingjing Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China
| | - Wenjing Yuan
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China
| | - Qingqing Shen
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China
| | - Qipeng Wu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China
| | - Zhenzhou Jiang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, PR China
| | - Wei Wu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China
| | - Luyong Zhang
- Center for Drug Screening and Pharmacodynamics Evaluation, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Xin Huang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing, PR China.,New Drug Screening Center, Institute of Pharmaceutical Research, China Pharmaceutical University, Nanjing, PR China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, PR China
| |
Collapse
|
13
|
Florenly F, Sugianto L, Lister INE, Girsang E, Ginting CN, Afifah E, Kusuma H, Rizal R, Widowati W. Protective Effect of Eugenol against Acetaminophen-Induced Hepatotoxicity in Human Hepatocellular Carcinoma Cells via Antioxidant, Anti-Inflammatory, and Anti-Necrotic Potency. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Overdoses acetaminophen (APAP) could cause acute liver failure, even though it used is for analgesics. APAP could cause hepatotoxicity due to multiple mediators of inflammation and oxidative stress. Eugenol has been reported to have anti-inflammatory and antioxidant activity but its hepatoprotective effect has not been widely reported.
AIM: The purpose of this research is to know if eugenol could protect HepG2 cells from APAP.
METHODS: HepG2 that induced by APAP as hepatotoxicity cells model was treated by using eugenol at 6.25 and 25 μg/mL. The protective effects of eugenol toward hepatotoxicity were evaluated by determine tumor necrosis factor-α (TNF-α) concentration, apoptotic activity, reactive oxygen species (ROS) level, also cytochrome (CYP)2E1 and GPX gene expression.
RESULTS: Eugenol at 6.25 and 25 μg/mL concentration can reduce TNF-α concentration, the apoptotic, necrotic, dead cells, and ROS level. Besides it can increase the gene expression (GPX and CYP2E1). The best hepatoprotective effect was found when using the eugenol at 25 μg/mL.
CONCLUSION: Therefore, eugenol can be used to protect HepG2 cells against APAP.
Collapse
|
14
|
Hwang JJ, Choi J, Rim YA, Nam Y, Ju JH. Application of Induced Pluripotent Stem Cells for Disease Modeling and 3D Model Construction: Focus on Osteoarthritis. Cells 2021; 10:cells10113032. [PMID: 34831254 PMCID: PMC8622662 DOI: 10.3390/cells10113032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
Since their discovery in 2006, induced pluripotent stem cells (iPSCs) have shown promising potential, specifically because of their accessibility and plasticity. Hence, the clinical applicability of iPSCs was investigated in various fields of research. However, only a few iPSC studies pertaining to osteoarthritis (OA) have been performed so far, despite the high prevalence rate of degenerative joint disease. In this review, we discuss some of the most recent applications of iPSCs in disease modeling and the construction of 3D models in various fields, specifically focusing on osteoarthritis and OA-related conditions. Notably, we comprehensively reviewed the successful results of iPSC-derived disease models in recapitulating OA phenotypes for both OA and early-onset OA to encompass their broad etiology. Moreover, the latest publications with protocols that have used iPSCs to construct 3D models in recapitulating various conditions, particularly the OA environment, were further discussed. With the overall optimistic results seen in both fields, iPSCs are expected to be more widely used for OA disease modeling and 3D model construction, which could further expand OA drug screening, risk assessment, and therapeutic capabilities.
Collapse
Affiliation(s)
- Joel Jihwan Hwang
- College of Public Health and Social Justice, Saint Louis University, St. Louis, MO 63103, USA;
| | - Jinhyeok Choi
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Yoojun Nam
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
| | - Ji Hyeon Ju
- YiPSCELL, Inc., 39 Banpo-daero, Seocho-gu, Seoul 06579, Korea; (J.C.); (Y.N.)
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
- Division of Rheumatology, Department of Internal Medicine, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul St. Mary’s Hospital, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
15
|
Identification of miR-199a-5p, miR-214-3p and miR-99b-5p as Fibrosis-Specific Extracellular Biomarkers and Promoters of HSC Activation. Int J Mol Sci 2021; 22:ijms22189799. [PMID: 34575957 PMCID: PMC8464755 DOI: 10.3390/ijms22189799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/27/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022] Open
Abstract
Liver fibrosis is characterized by the accumulation of extracellular matrix (ECM) resulting in the formation of fibrous scars. In the clinic, liver biopsies are the standard diagnostic method despite the potential for clinical complications. miRNAs are single-stranded, non-coding RNAs that can be detected in tissues, body fluids and cultured cells. The regulation of many miRNAs has been linked to tissue damage, including liver fibrosis in patients, resulting in aberrant miRNA expression/release. Experimental evidence also suggests that miRNAs are regulated in a similar manner in vitro and could thus serve as translational in vitro–in vivo biomarkers. In this work, we set out to identify and characterize biomarkers for liver fibrosis that could be used in vitro and clinically for research and diagnostic purposes. We focused on miRNAs released from hepatic 3D cultures exposed to methotrexate (MTX), which causes fibrosis, and acetaminophen (APAP), an acute hepatotoxicant with no clinically relevant association to liver fibrosis. Using a 3D in vitro model, we corroborated compound-specific responses as we show MTX induced a fibrotic response, and APAP did not. Performing miRNA-seq of cell culture supernatants, we identified potential miRNA biomarkers (miR-199a-5p, miR-214-3p, niRNA-125a-5p and miR-99b-5p) that were associated with a fibrotic phenotype and not with hepatocellular damage alone. Moreover, transfection of HSC with miR-199a-5p led to decreased expression of caveolin-1 and increased α-SMA expression, suggesting its role in HSC activation. In conclusion, we propose that extracellular miR-214-3p, miR-99b-5p, miR-125a-5p and specifically miR-199a-5p could contribute towards a panel of miRNAs for identifying liver fibrosis and that miR-199a-5p, miR-214-3p and miR-99b-5p are promoters of HSC activation.
Collapse
|
16
|
Zhang Y, Liu H, Lin X, Zhang F, Meng P, Tan S, Lammi MJ, Guo X. Dysregulation of Cells Cycle and Apoptosis in Human Induced Pluripotent Stem Cells Chondrocytes Through p53 Pathway by HT-2 Toxin: An in vitro Study. Front Genet 2021; 12:677723. [PMID: 34421989 PMCID: PMC8371750 DOI: 10.3389/fgene.2021.677723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/28/2021] [Indexed: 01/23/2023] Open
Abstract
Kashin–Beck disease (KBD) mainly damages growth plate of adolescents and is susceptible to both gene and gene–environmental risk factors. HT-2 toxin, which is a primary metabolite of T-2 toxin, was regarded as one of the environmental risk factors of KBD. We used successfully generated KBD human induced pluripotent stem cells (hiPSCs) and control hiPSCs, which carry different genetic information. They have potential significance in exploring the effects of HT-2 toxin on hiPSC chondrocytes and interactive genes with HT-2 toxin for the purpose of providing a cellular disease model for KBD. In this study, we gave HT-2 toxin treatment to differentiating hiPSC chondrocytes in order to investigate the different responses of KBD hiPSC chondrocytes and control hiPSC chondrocytes to HT-2 toxin. The morphology of HT-2 toxin-treated hiPSC chondrocytes investigated by transmission electron microscope clearly showed that the ultrastructure of organelles was damaged and type II collagen expression in hiPSC chondrocytes was downregulated by HT-2 treatment. Moreover, dysregulation of cell cycle was observed; and p53, p21, and CKD6 gene expressions were dysregulated in hiPSC chondrocytes after T-2 toxin treatment. Flow cytometry also demonstrated that there were significantly increased amounts of late apoptotic cells in KBD hiPSC chondrocytes and that the mRNA expression level of Fas was upregulated. In addition, KBD hiPSC chondrocytes presented stronger responses to HT-2 toxin than control hiPSC chondrocytes. These findings confirmed that HT-2 is an environmental risk factor of KBD and that p53 pathway interacted with HT-2 toxin, causing damaged ultrastructure of organelles, accelerating cell cycle in G1 phase, and increasing late apoptosis in KBD hiPSC chondrocytes.
Collapse
Affiliation(s)
- Yanan Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Huan Liu
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Xialu Lin
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Feng'e Zhang
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Peilin Meng
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Sijia Tan
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| | - Mikko J Lammi
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Department of Integrative Medical Biology, University of Umeå, Umeå, Sweden
| | - Xiong Guo
- School of Public Health, Health Science Center of Xi'an Jiaotong University, Xi'an, China.,Key Laboratory of Trace Elements and Endemic Diseases of National Health Commission and Collaborative Innovation Center of Endemic Diseases and Health Promotion in Silk Road Region, Xi'an, China
| |
Collapse
|
17
|
Jin M, Yi X, Liao W, Chen Q, Yang W, Li Y, Li S, Gao Y, Peng Q, Zhou S. Advancements in stem cell-derived hepatocyte-like cell models for hepatotoxicity testing. Stem Cell Res Ther 2021; 12:84. [PMID: 33494782 PMCID: PMC7836452 DOI: 10.1186/s13287-021-02152-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced liver injury (DILI) is one of the leading causes of clinical trial failures and high drug attrition rates. Currently, the commonly used hepatocyte models include primary human hepatocytes (PHHs), animal models, and hepatic cell lines. However, these models have disadvantages that include species-specific differences or inconvenient cell extraction methods. Therefore, a novel, inexpensive, efficient, and accurate model that can be applied to drug screening is urgently needed. Owing to their self-renewable ability, source abundance, and multipotent competence, stem cells are stable sources of drug hepatotoxicity screening models. Because 3D culture can mimic the in vivo microenvironment more accurately than can 2D culture, the former is commonly used for hepatocyte culture and drug screening. In this review, we introduce the different sources of stem cells used to generate hepatocyte-like cells and the models for hepatotoxicity testing that use stem cell-derived hepatocyte-like cells.
Collapse
Affiliation(s)
- Meixian Jin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Xiao Yi
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Liao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qi Chen
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China
| | - Wanren Yang
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yang Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shao Li
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yi Gao
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Qing Peng
- General Surgery Center, Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.
| | - Shuqin Zhou
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, China.
| |
Collapse
|
18
|
Chen J, Wang W, Jiang M, Yang M, Wei W. Combination therapy of ginsenoside compound K and methotrexate was efficient in elimination of anaemia and reduction of disease activity in adjuvant-induced arthritis rats. PHARMACEUTICAL BIOLOGY 2020; 58:1131-1139. [PMID: 33198544 PMCID: PMC7671656 DOI: 10.1080/13880209.2020.1844761] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
CONTEXT Ginsenoside compound K (CK) has anti-inflammatory, immunoregulatory, and myelosuppressive protective effects. Methotrexate (MTX) is widely used in combination therapy for rheumatoid arthritis (RA). OBJECTIVE To evaluate the effects of combination therapy of CK and MTX on anaemia and anti-arthritis in adjuvant-induced arthritis (AA) rats. MATERIALS AND METHODS AA was induced in rats by Complete Freund's adjuvant, and divided into five groups (n = 10): normal, AA, CK 80 mg/kg, combination therapy (80 mg/kg CK combined with 0.5 mg/kg MTX), and MTX 0.5 mg/kg. From day 12, CK (once a day for 15 days) or MTX (once every 3 days, five times) were intragastrically administered. RESULTS Combination therapy showed increased haemoglobin to 148.5 ± 10.1 g/L compared with AA (129.8 ± 11.7 g/L) and MTX (128.8 ± 18.4 g/L), and decreased reticulocytes in peripheral blood to 4.9 ± 1.1% compared with MTX (9.3 ± 3.3%). In combination therapy group, paw swelling decreased to 5.6 ± 4.3 mL compared with CK (9.4 ± 3.9 mL) and MTX (13.5 ± 7.4 mL), and swollen joint count decreased to 1.4 ± 0.8 compared with CK (2.1 ± 1.0) and MTX (2.4 ± 1.2) at day 24. Combination therapy showed decreased IL-6 to 25.1 ± 17.2 pg/mL compared with MTX (44.9 ± 4.8 pg/mL), and decreased IL-17 to 5.8 ± 3.9 pg/mL compared with MTX (10.7 ± 4.2 pg/mL). CONCLUSION The anti-anaemia effect of CK deserves further study, and CK can be a candidate effective drug for combined treatment in RA with anaemia.
Collapse
Affiliation(s)
- Jingyu Chen
- Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
- Jingyu Chen Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wu Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| | - Mengya Jiang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| | - Mei Yang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, China
- CONTACT Wei Wei
| |
Collapse
|
19
|
Shahzad K, Mushtaq S, Rizwan M, Khalid W, Atif M, Din FU, Ahmad N, Abbasi R, Ali Z. Field-controlled magnetoelectric core-shell CoFe 2O 4@BaTiO 3 nanoparticles as effective drug carriers and drug release in vitro. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111444. [PMID: 33321584 DOI: 10.1016/j.msec.2020.111444] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/16/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022]
Abstract
The targeted drug release at tumor cells while sparing normal cells is a huge challenge. Core-shell magnetoelectric (ME) nanoparticles have addressed this problem using shape-dependent magneto-electric attributes. The colloidally stable, core-shell cobalt ferrite@barium titanate (CFO@BTO) ME nanoparticles (NPs) used for in vitro study were synthesized using sonochemical method. The structural characteristics and core-shell morphology were analyzed by X-ray Diffraction (XRD) and Transmission Electron Microscopy (TEM) respectively. Further magnetic and exchange coupling between two phases of ME nanostructures were studied at room temperature. Colloidal stability was studied in different suspension solutions (Water, SBB, PBS, and DMEM) using dynamic light scattering. Subsequently, the synthesized nanoparticles were functionalized with anticancer drugs including doxorubicin and methotrexate up to 80% via (EDC) chemistry. In vitro cytotoxicity studies carried out on human hepatocellular carcinoma (HepG2) and human malignant melanoma (HT144), cells validated the magneto-electric property of CFO@BTO nano-carriers in the presence of external magnetic field (5 mT), with significantly enhanced cytotoxicity when compared to free drugs and without field replicates. The resulted IC50 values ranging from 5.3-7.3 μg/ml compared to 30.1-43.1 μg/ml in the absence of a magnetic field also confirmed the involved physical attributes of magnetoelectric nanostructures. The fluorescent microscopy results also indicated the increased apoptosis in magnetic field-assisted samples. Finally, hemolysis assay indicated the suitability of CFO@BTO nano-carriers for intravenous applications at IC50 concentration.
Collapse
Affiliation(s)
- Khuram Shahzad
- Functional Materials Lab, Department of Physics, Air University Sector E-9, Islamabad, Pakistan
| | - Sadaf Mushtaq
- Department of Biotechnology, Quaid-i-Azam University Islamabad, Pakistan; Institute of Biomedical and Genetic Engineering, 24 Mauve Area, Sector G-9/1, Islamabad, Pakistan
| | - Muhammad Rizwan
- Functional Materials Lab, Department of Physics, Air University Sector E-9, Islamabad, Pakistan
| | - Waqas Khalid
- Functional Materials Lab, Department of Physics, Air University Sector E-9, Islamabad, Pakistan
| | - Muhammad Atif
- Functional Materials Lab, Department of Physics, Air University Sector E-9, Islamabad, Pakistan
| | - Fakhar Ud Din
- Nanomedicine Research Group, Department of Pharmacy, Quaid-i-Azam University Islamabad, 45320, Pakistan
| | - Nafees Ahmad
- Institute of Biomedical and Genetic Engineering, 24 Mauve Area, Sector G-9/1, Islamabad, Pakistan
| | - Rashda Abbasi
- Institute of Biomedical and Genetic Engineering, 24 Mauve Area, Sector G-9/1, Islamabad, Pakistan
| | - Zulqurnain Ali
- Functional Materials Lab, Department of Physics, Air University Sector E-9, Islamabad, Pakistan.
| |
Collapse
|
20
|
Analysis of association of ADORA 2A and ADORA 3 polymorphisms genotypes/haplotypes with efficacy and toxicity of methotrexate in patients with Rheumatoid arthritis. THE PHARMACOGENOMICS JOURNAL 2020; 20:784-791. [PMID: 32448869 DOI: 10.1038/s41397-020-0168-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
Adenosine receptors ADORA2A and ADORA3 are part of the adenosine-mediated antiinflammatory pathway and are overexpressed in patients with Rheumatoid arthritis (RA). Methotrexate (MTX) antiinflammatory effects are partially mediated via increased release of adenosine into extracellular space. Polymorphisms in ADORA2A and ADORA3 genes may have an impact on the efficacy and toxicity of MTX in RA patients. The study included 127 RA patients. Treatment efficacy was estimated using the changes in Disease activity score (DAS28) after 6 months of MTX monotherapy, according to EULAR response criteria. Patients with good and moderate response were classified as "responders", and with a poor response as "nonresponders". Adverse effects were collected during the follow-up period. Genotyping for polymorphisms within ADORA2A gene (rs2298383, rs2236624, rs5751876, rs17004921) and ADORA3 gene (rs2298191, rs1544223, rs3393) was performed using the KASPar assays. Among patients 112 (88.19%) were responders (18.8% good, 81.2% moderate). We observed no association between analyzed genotypes or alleles and MTX response by EULAR criteria but carriers of ADORA2A rs17004921 T allele (CT + TT) had a higher DAS28 decrease after 6 months of treatment than patients with CC genotype (p = 0.013). Adverse effects were reported in 31 patients (24.41%). Bone erosions were present in 82 (64.6%) patients. Haplotype block was observed among all 3 analyzed polymorphisms within ADORA3 gene and TAA haplotype was associated with bone erosions (29% vs 15.6%, p = 0.023) and hepatotoxicity (51.3% vs 21.6%, p = 0.013). According to our study, ADORA3 TAA haplotype may be associated with bone erosions and hepatotoxicity in RA patients treated with MTX.
Collapse
|
21
|
Zhang X, Jiang T, Chen D, Wang Q, Zhang LW. Three-dimensional liver models: state of the art and their application for hepatotoxicity evaluation. Crit Rev Toxicol 2020; 50:279-309. [DOI: 10.1080/10408444.2020.1756219] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Xihui Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Tianyan Jiang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Dandan Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| | - Qi Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control (NIFDC), China Food and Drug Administration (CFDA), Beijing, P. R. China
| | - Leshuai W. Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, P. R. China
| |
Collapse
|