1
|
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
|
2
|
Tong T, Hao C, Shen J, Liu S, Yan S, Aslam MS, Chen Y, Chen W, Li J, Li Y, Zeng J, Li M, You Z, Gulizhaerkezi T, Wei S, Zhu A, Meng X. Electroacupuncture ameliorates chronic unpredictable mild stress-induced depression-like behavior and cognitive impairment through suppressing oxidative stress and neuroinflammation in rats. Brain Res Bull 2024; 206:110838. [PMID: 38123022 DOI: 10.1016/j.brainresbull.2023.110838] [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: 07/28/2023] [Revised: 11/01/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Depression is associated with lowered mood, anxiety, anhedonia, cognitive impairments, and even suicidal tendencies in severe cases. Yet few studies have directed acupuncture's mechanism toward enhancing axonal repair correlated with synaptic plasticity and anti-inflammatory effects related to oxidative stress in the hippocampus. METHODS Male Sprague-Dawley (SD) rats were randomly divided into control group (CON), chronic unpredictable mild stress (CUMS) group, CUMS + electroacupuncture group (EA), and CUMS + fluoxetine group (FLX) (n = 10/group). Rats were given a 28-day treatment at the Shangxing (GV23) and Fengfu (GV16) acupoints with electroacupuncture or fluoxetine (2.1 mg/kg). RESULTS Rats exposed to CUMS induced depression-like behaviors and spatial learning-memory impairment, changed the ionized calcium binding adaptor molecule 1 (IBA-1), Vglut1, myelin basic protein (MBP), and postsynaptic density protein 95 (PSD95) level of hippocampal, increased the Nod-like receptor protein 3 (NLRP3), atypical squamous cell (ASC), Caspase level and hippocampal reactive oxygen species (ROS), and prompted the activation of Epha4-mediated signaling and an inflammatory response. Conversely, electroacupuncture administration reduced these changes and prevented depression-like behaviors and cognitive impairment. Electroacupuncture also promoted hippocampal expression of Sirtuin1(SIRT1), Nuclear factor erythroid 2-like (Nrf2), Heme oxygenase-1 (HO-1); reduced the expression of interleukin-1β (IL-1β), interleukin-18 (IL-18), and tumor necrosis factor-alpha (TNF-α); and prevented neural damage, particularly the synaptic myelin sheath, and neuroinflammation by regulating Eph receptor A4 (EphA4) in the hippocampal. CONCLUSION These results indicate that electroacupuncture prevents depression-like behaviors with cognitive impairment and synaptic and neuronal damage, probably by reducing EphA4, which mediates ROS hyperfunction and the inflammatory response.
Collapse
Affiliation(s)
- Tao Tong
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Chongyao Hao
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Junliang Shen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Siyu Liu
- Longyan Hospital of Traditional Chinese Medicine of Xiamen University, Longyan, Fujian, PR China.
| | - Simin Yan
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | | | - Yiping Chen
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Wenjie Chen
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Jianguo Li
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Yuhan Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, PR China.
| | - Jingyu Zeng
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Meng Li
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Zhuoran You
- Second Clinical College, Shanxi University of Traditional Chinese Medicine, Taiyuan, Shanxi, PR China.
| | - Tuergong Gulizhaerkezi
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Simiao Wei
- Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| | - Anning Zhu
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xianjun Meng
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Department of Traditional Chinese Medicine, School of Medicine, Xiamen University, Xiamen, Fujian, PR China.
| |
Collapse
|
3
|
Zhou HY, Luo Q, Sui H, Du XN, Zhao YJ, Liu L, Guan Q, Zhou Y, Wen QS, Shi Y, Sun Y, Lin HL, Wang DP. Recent advances in the involvement of epigenetics in the pathogenesis of systemic lupus erythematosus. Clin Immunol 2024; 258:109857. [PMID: 38043757 DOI: 10.1016/j.clim.2023.109857] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/27/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Systemic lupus erythematosus (SLE) is a typical systemic autoimmune disease that manifests as skin rash, arthritis, lymphadenopathy, and multiple organ lesions. Epigenetics, including DNA methylation, histone modification, and non-coding RNA regulation, mainly affect the function and characteristics of cells through the regulation of gene transcription or translation. Increasing evidence indicates that there are a variety of complex epigenetic effects in patients with SLE, which interfere with the differentiation and function of T, and B lymphocytes, monocytes, and neutrophils, and enhance the expression of SLE-associated pathogenic genes. This paper summarizes our currently knowledge regarding pathogenesis of SLE, and introduces current advances in the epigenetic regulation of SLE from three aspects: immune function, inflammatory response, and lupus complications. We propose that epigenetic changes could be used as potential biomarkers and therapeutic targets of SLE.
Collapse
Affiliation(s)
- Hong-Yan Zhou
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi Luo
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hua Sui
- Integrated TCM and Western Medicine Collage of Dalian Medical University, Dalian, China
| | - Xiang-Ning Du
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang-Jianing Zhao
- Integrated TCM and Western Medicine Collage of Dalian Medical University, Dalian, China
| | - Lu Liu
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qing Guan
- Integrated TCM and Western Medicine Collage of Dalian Medical University, Dalian, China
| | - Yue Zhou
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qing-Si Wen
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yan Shi
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Sun
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong-Li Lin
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Da-Peng Wang
- Nephrology Department of First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
4
|
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
|
5
|
Lucchino V, Scaramuzzino L, Scalise S, Lo Conte M, Zannino C, Benedetto GL, Aguglia U, Ferlazzo E, Cuda G, Parrotta EI. Insights into the Genetic Profile of Two Siblings Affected by Unverricht-Lundborg Disease Using Patient-Derived hiPSCs. Cells 2022; 11:3491. [PMID: 36359887 PMCID: PMC9655992 DOI: 10.3390/cells11213491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/29/2023] Open
Abstract
Unverricht-Lundborg disease (ULD), also known as progressive myoclonic epilepsy 1 (EPM1), is a rare autosomal recessive neurodegenerative disorder characterized by a complex symptomatology that includes action- and stimulus-sensitive myoclonus and tonic-clonic seizures. The main cause of the onset and development of ULD is a repeat expansion of a dodecamer sequence localized in the promoter region of the gene encoding cystatin B (CSTB), an inhibitor of lysosomal proteases. Although this is the predominant mutation found in most patients, the physio-pathological mechanisms underlying the disease complexity remain largely unknown. In this work, we used patient-specific iPSCs and their neuronal derivatives to gain insight into the molecular and genetic machinery responsible for the disease in two Italian siblings affected by different phenotypes of ULD. Specifically, fragment length analysis on amplified CSTB promoters found homozygous status for dodecamer expansion in both patients and showed that the number of dodecamer repeats is the same in both. Furthermore, the luciferase reporter assay showed that the CSTB promoter activity was similarly reduced in both lines compared to the control. This information allowed us to draw important conclusions: (1) the phenotypic differences of the patients do not seem to be strictly dependent on the genetic mutation around the CSTB gene, and (2) that some other molecular mechanisms, not yet clearly identified, might be taken into account. In line with the inhibitory role of cystatin B on cathepsins, molecular investigations performed on iPSCs-derived neurons showed an increased expression of lysosomal cathepsins (B, D, and L) and a reduced expression of CSTB protein. Intriguingly, the increase in cathepsin expression does not appear to be correlated with the residual amount of CSTB, suggesting that other mechanisms, in addition to the regulation of cathepsins, could be involved in the pathological complexity of the disease.
Collapse
Affiliation(s)
- Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Luana Scaramuzzino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giorgia Lucia Benedetto
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | | |
Collapse
|
6
|
Scalise S, Zannino C, Lucchino V, Lo Conte M, Scaramuzzino L, Cifelli P, D’Andrea T, Martinello K, Fucile S, Palma E, Gambardella A, Ruffolo G, Cuda G, Parrotta EI. Human iPSC Modeling of Genetic Febrile Seizure Reveals Aberrant Molecular and Physiological Features Underlying an Impaired Neuronal Activity. Biomedicines 2022; 10:biomedicines10051075. [PMID: 35625812 PMCID: PMC9138645 DOI: 10.3390/biomedicines10051075] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Mutations in SCN1A gene, encoding the voltage-gated sodium channel (VGSC) NaV1.1, are widely recognized as a leading cause of genetic febrile seizures (FS), due to the decrease in the Na+ current density, mainly affecting the inhibitory neuronal transmission. Here, we generated induced pluripotent stem cells (iPSCs)-derived neurons (idNs) from a patient belonging to a genetically well-characterized Italian family, carrying the c.434T > C mutation in SCN1A gene (hereafter SCN1AM145T). A side-by-side comparison of diseased and healthy idNs revealed an overall maturation delay of SCN1AM145T cells. Membranes isolated from both diseased and control idNs were injected into Xenopus oocytes and both GABA and AMPA currents were successfully recorded. Patch-clamp measurements on idNs revealed depolarized action potential for SCN1AM145T, suggesting a reduced excitability. Expression analyses of VGSCs and chloride co-transporters NKCC1 and KCC2 showed a cellular “dysmaturity” of mutated idNs, strengthened by the high expression of SCN3A, a more fetal-like VGSC isoform, and a high NKCC1/KCC2 ratio, in mutated cells. Overall, we provide strong evidence for an intrinsic cellular immaturity, underscoring the role of mutant NaV1.1 in the development of FS. Furthermore, our data are strengthening previous findings obtained using transfected cells and recordings on human slices, demonstrating that diseased idNs represent a powerful tool for personalized therapy and ex vivo drug screening for human epileptic disorders.
Collapse
Affiliation(s)
- Stefania Scalise
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Clara Zannino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Valeria Lucchino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Luana Scaramuzzino
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
| | - Pierangelo Cifelli
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of Aquila, 67100 Aquila, Italy;
| | - Tiziano D’Andrea
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
| | | | - Sergio Fucile
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
- IRCCS Neuromed, Via Atinense, 86077 Pozzilli, Italy;
| | - Eleonora Palma
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
| | - Antonio Gambardella
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.G.); (E.I.P.)
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, University of Rome, Sapienza, P.le Aldo Moro, 5, 00185 Rome, Italy; (T.D.); (S.F.); (E.P.)
- IRCCS San Raffaele Roma, Via della Pisana, 00163 Rome, Italy
- Correspondence: (G.R.); (G.C.)
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (S.S.); (C.Z.); (V.L.); (M.L.C.); (L.S.)
- Correspondence: (G.R.); (G.C.)
| | - Elvira Immacolata Parrotta
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Viale Europa, 88100 Catanzaro, Italy; (A.G.); (E.I.P.)
| |
Collapse
|
7
|
Vitronectin, a Novel Urinary Proteomic Biomarker, Promotes Cell Pyroptosis in Juvenile Systemic Lupus Erythematosus. Mediators Inflamm 2022; 2022:8447675. [PMID: 35462789 PMCID: PMC9020974 DOI: 10.1155/2022/8447675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 02/09/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Identifying new markers of juvenile systemic lupus erythematosus (JSLE) is critical event to predict patient stratification and prognosis. The aim of the present study is to analyze alteration of urinary protein expression and screen potential valuable biomarkers in juvenile systemic lupus erythematosus (JSLE). Methods The urine was collected from the patients with or without JSLE and detected by mass spectrometry to analyze proteomic changes. ELISA was used to verify the Vitronectin (VTN) changes in a new set of patients. The clinical correlation was performed to analyze between VTN and clinical pathological parameters. WB and ELISA were used to analyze VTN-mediated cell pyroptosis. Results Herein, we have identified a group of 105 differentially expressed proteins with ≥1.3-fold upregulation or ≤0.77-fold downregulation in JSLE patients. These proteins were involved in several important biological processes, including acute phase inflammatory responses, complement activation, hemostasis, and immune system regulation through Gene Ontology and functional enrichment analysis. Interestingly, urinary ephrin type-A receptor 4 (EPHA4) and VTN were significantly reduced in both inactive and active JSLE patients, and VTN treatment in THP-1 derived macrophages led to a significant increased cell pyroptosis by activation of Nod-like receptor family protein 3 (NLRP3) inflammasomes, resulting in caspase-1 activation, cleaved gasdermin D (GSDMD), and IL-18 secretion. Most importantly, the urinary VTN was also linearly correlated with clinical characteristics of JSLE, implying that VTN could be a specific diagnostic biomarker to distinguish inactive and active JSLE. Conclusion This study provided a novel role of VTN in pyroptosis in JSLE through the urinary proteomic profile for JSLE, which could be a nonintrusive monitoring strategy in clinical diagnosis.
Collapse
|
8
|
Zhao X, Ge L, Wang J, Song Z, Ni B, He X, Ruan Z, You Y. Exploration of Potential Integrated Models of N6-Methyladenosine Immunity in Systemic Lupus Erythematosus by Bioinformatic Analyses. Front Immunol 2022; 12:752736. [PMID: 35197962 PMCID: PMC8859446 DOI: 10.3389/fimmu.2021.752736] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 12/31/2021] [Indexed: 01/27/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is a prototypical systemic autoimmune disease of unknown etiology. The epigenetic regulation of N6-methyladenosine (m6A) modification in immunity is emerging. However, few studies have focused on SLE and m6A immune regulation. In this study, we aimed to explore a potential integrated model of m6A immunity in SLE. The models were constructed based on RNA-seq data of SLE. A consensus clustering algorithm was applied to reveal the m6A-immune signature using principal component analysis (PCA). Univariate and multivariate Cox regression analyses and Kaplan–Meier analysis were used to evaluate diagnostic differences between groups. The effects of m6A immune-related characteristics were investigated, including risk evaluation of m6A immune phenotype-related characteristics, immune cell infiltration profiles, diagnostic value, and enrichment pathways. CIBERSORT, ESTIMATE, and single-sample gene set enrichment analysis (ssGSEA) were used to evaluate the relative immune cell infiltrations (ICIs) of the samples. Conventional bioinformatics methods were used to identify key m6A regulators, pathways, gene modules, and the coexpression network of SLE. In summary, our study revealed that IGFBP3 (as a key m6A regulator) and two pivotal immune genes (CD14 and IDO1) may aid in the diagnosis and treatment of SLE. The potential integrated models of m6A immunity that we developed could guide clinical management and may contribute to the development of personalized immunotherapy strategies.
Collapse
Affiliation(s)
- Xingwang Zhao
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lan Ge
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Juan Wang
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhiqiang Song
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Ni
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaochong He
- Department of Nursing Administration, Faculty of Nursing, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yi You, ; Xiaochong He, ; Zhihua Ruan,
| | - Zhihua Ruan
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yi You, ; Xiaochong He, ; Zhihua Ruan,
| | - Yi You
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Yi You, ; Xiaochong He, ; Zhihua Ruan,
| |
Collapse
|
9
|
Smith AST, Chun C, Hesson J, Mathieu J, Valdmanis PN, Mack DL, Choi BO, Kim DH, Bothwell M. Human Induced Pluripotent Stem Cell-Derived TDP-43 Mutant Neurons Exhibit Consistent Functional Phenotypes Across Multiple Gene Edited Lines Despite Transcriptomic and Splicing Discrepancies. Front Cell Dev Biol 2021; 9:728707. [PMID: 34660586 PMCID: PMC8511491 DOI: 10.3389/fcell.2021.728707] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/06/2021] [Indexed: 11/25/2022] Open
Abstract
Gene editing technologies hold great potential to enhance our ability to model inheritable neurodegenerative diseases. Specifically, engineering multiple amyotrophic lateral sclerosis (ALS) mutations into isogenic cell populations facilitates determination of whether different causal mutations cause pathology via shared mechanisms, and provides the capacity to separate these mechanisms from genotype-specific effects. As gene-edited, cell-based models of human disease become more commonplace, there is an urgent need to verify that these models constitute consistent and accurate representations of native biology. Here, commercially sourced, induced pluripotent stem cell-derived motor neurons from Cellular Dynamics International, edited to express the ALS-relevant mutations TDP-43M337V and TDP-43Q331K were compared with in-house derived lines engineered to express the TDP-43Q331K mutation within the WTC11 background. Our results highlight electrophysiological and mitochondrial deficits in these edited cells that correlate with patient-derived cells, suggesting a consistent cellular phenotype arising from TDP-43 mutation. However, significant differences in the transcriptomic profiles and splicing behavior of the edited cells underscores the need for careful comparison of multiple lines when attempting to use these cells as a means to better understand the onset and progression of ALS in humans.
Collapse
Affiliation(s)
- Alec S T Smith
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| | - Changho Chun
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Jennifer Hesson
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States.,Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Julie Mathieu
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States.,Department of Comparative Medicine, University of Washington, Seattle, WA, United States
| | - Paul N Valdmanis
- Division of Medical Genetics, University of Washington, Seattle, WA, United States
| | - David L Mack
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States.,Department of Bioengineering, University of Washington, Seattle, WA, United States.,Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Stem Cell and Regenerative Medicine Institute, Samsung Medical Center, Seoul, South Korea.,Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, South Korea
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA, United States.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States.,Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark Bothwell
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
10
|
Scaramuzzino L, Lucchino V, Scalise S, Lo Conte M, Zannino C, Sacco A, Biamonte F, Parrotta EI, Costanzo FS, Cuda G. Uncovering the Metabolic and Stress Responses of Human Embryonic Stem Cells to FTH1 Gene Silencing. Cells 2021; 10:2431. [PMID: 34572080 PMCID: PMC8469604 DOI: 10.3390/cells10092431] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/03/2021] [Accepted: 09/13/2021] [Indexed: 12/22/2022] Open
Abstract
Embryonic stem cells (ESCs) are pluripotent cells with indefinite self-renewal ability and differentiation properties. To function properly and maintain genomic stability, ESCs need to be endowed with an efficient repair system as well as effective redox homeostasis. In this study, we investigated different aspects involved in ESCs' response to iron accumulation following stable knockdown of the ferritin heavy chain (FTH1) gene, which encodes for a major iron storage protein with ferroxidase activity. Experimental findings highlight unexpected and, to a certain extent, paradoxical results. If on one hand FTH1 silencing does not correlate with increased ROS production nor with changes in the redox status, strengthening the concept that hESCs are extremely resistant and, to a certain extent, even refractory to intracellular iron imbalance, on the other, the differentiation potential of hESCs seems to be affected and apoptosis is observed. Interestingly, we found that FTH1 silencing is accompanied by a significant activation of the nuclear factor (erythroid-derived-2)-like 2 (Nrf2) signaling pathway and pentose phosphate pathway (PPP), which crosstalk in driving hESCs antioxidant cascade events. These findings shed new light on how hESCs perform under oxidative stress, dissecting the molecular mechanisms through which Nrf2, in combination with PPP, counteracts oxidative injury triggered by FTH1 knockdown.
Collapse
Affiliation(s)
- Luana Scaramuzzino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Valeria Lucchino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Stefania Scalise
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Michela Lo Conte
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Clara Zannino
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Alessandro Sacco
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| | - Flavia Biamonte
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
- Center of Interdepartmental Services (CIS), “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | | | - Francesco Saverio Costanzo
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
- Center of Interdepartmental Services (CIS), “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy
| | - Giovanni Cuda
- Research Centre for Advanced Biochemistry and Molecular Biology, Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy; (L.S.); (V.L.); (S.S.); (M.L.C.); (C.Z.); (A.S.); (F.B.); (F.S.C.); (G.C.)
| |
Collapse
|
11
|
Deciphering the Role of Wnt and Rho Signaling Pathway in iPSC-Derived ARVC Cardiomyocytes by In Silico Mathematical Modeling. Int J Mol Sci 2021; 22:ijms22042004. [PMID: 33670616 PMCID: PMC7923182 DOI: 10.3390/ijms22042004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic Right Ventricular cardiomyopathy (ARVC) is an inherited cardiac muscle disease linked to genetic deficiency in components of the desmosomes. The disease is characterized by progressive fibro-fatty replacement of the right ventricle, which acts as a substrate for arrhythmias and sudden cardiac death. The molecular mechanisms underpinning ARVC are largely unknown. Here we propose a mathematical model for investigating the molecular dynamics underlying heart remodeling and the loss of cardiac myocytes identity during ARVC. Our methodology is based on three computational models: firstly, in the context of the Wnt pathway, we examined two different competition mechanisms between β-catenin and Plakoglobin (PG) and their role in the expression of adipogenic program. Secondly, we investigated the role of RhoA-ROCK pathway in ARVC pathogenesis, and thirdly we analyzed the interplay between Wnt and RhoA-ROCK pathways in the context of the ARVC phenotype. We conclude with the following remark: both Wnt/β-catenin and RhoA-ROCK pathways must be inactive for a significant increase of PPARγ expression, suggesting that a crosstalk mechanism might be responsible for mediating ARVC pathogenesis.
Collapse
|
12
|
Parrotta EI, Scalise S, Scaramuzzino L, Cuda G. Stem Cells: The Game Changers of Human Cardiac Disease Modelling and Regenerative Medicine. Int J Mol Sci 2019; 20:E5760. [PMID: 31744081 PMCID: PMC6888119 DOI: 10.3390/ijms20225760] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022] Open
Abstract
A comprehensive understanding of the molecular basis and mechanisms underlying cardiac diseases is mandatory for the development of new and effective therapeutic strategies. The lack of appropriate in vitro cell models that faithfully mirror the human disease phenotypes has hampered the understanding of molecular insights responsible of heart injury and disease development. Over the past decade, important scientific advances have revolutionized the field of stem cell biology through the remarkable discovery of reprogramming somatic cells into induced pluripotent stem cells (iPSCs). These advances allowed to achieve the long-standing ambition of modelling human disease in a dish and, more interestingly, paved the way for unprecedented opportunities to translate bench discoveries into new therapies and to come closer to a real and effective stem cell-based medicine. The possibility to generate patient-specific iPSCs, together with the new advances in stem cell differentiation procedures and the availability of novel gene editing approaches and tissue engineering, has proven to be a powerful combination for the generation of phenotypically complex, pluripotent stem cell-based cellular disease models with potential use for early diagnosis, drug screening, and personalized therapy. This review will focus on recent progress and future outcome of iPSCs technology toward a customized medicine and new therapeutic options.
Collapse
Affiliation(s)
- Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, Research Center for Advanced Biochemistry and Molecular Biology, University “Magna Graecia” of Catanzaro, 88100 Loc., Germaneto, Catanzaro, Italy; (S.S.); (L.S.); (G.C.)
| | | | | | | |
Collapse
|
13
|
De Angelis MT, Santamaria G, Parrotta EI, Scalise S, Lo Conte M, Gasparini S, Ferlazzo E, Aguglia U, Ciampi C, Sgura A, Cuda G. Establishment and characterization of induced pluripotent stem cells (iPSCs) from central nervous system lupus erythematosus. J Cell Mol Med 2019; 23:7382-7394. [PMID: 31536674 PMCID: PMC6815917 DOI: 10.1111/jcmm.14598] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 08/02/2019] [Accepted: 08/06/2019] [Indexed: 12/16/2022] Open
Abstract
Involvement of the central nervous system (CNS) is an uncommon feature in systemic lupus erythematosus (SLE), making diagnosis rather difficult and challenging due to the poor specificity of neuropathic symptoms and neurological symptoms. In this work, we used human‐induced pluripotent stem cells (hiPSCs) derived from CNS‐SLE patient, with the aim to dissect the molecular insights underlying the disease by gene expression analysis and modulation of implicated pathways. CNS‐SLE‐derived hiPSCs allowed us to provide evidence of Erk and Akt pathways involvement and to identify a novel cohort of potential biomarkers, namely CHCHD2, IDO1, S100A10, EPHA4 and LEFTY1, never reported so far. We further extended the study analysing a panel of oxidative stress‐related miRNAs and demonstrated, under normal or stress conditions, a strong dysregulation of several miRNAs in CNS‐SLE‐derived compared to control hiPSCs. In conclusion, we provide evidence that iPSCs reprogrammed from CNS‐SLE patient are a powerful useful tool to investigate the molecular mechanisms underlying the disease and to eventually develop innovative therapeutic approaches.
Collapse
Affiliation(s)
- Maria Teresa De Angelis
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Elvira Immacolata Parrotta
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Stefania Scalise
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Michela Lo Conte
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| | - Sara Gasparini
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Edoardo Ferlazzo
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Umberto Aguglia
- Department of Medical and Surgical Sciences, "Magna Graecia" University, Catanzaro, Italy.,Regional Epilepsy Centre, Great Metropolitan Hospital, Reggio Calabria, Italy
| | - Clara Ciampi
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Antonella Sgura
- Department of Science, University of Rome " Roma Tre", Rome, Italy
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Stem Cell Laboratory, Research Center for Advanced Biochemistry and Molecular Biology, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|