1
|
Li W, Zhao X, Ren C, Gao S, Han Q, Lu M, Li X. The therapeutic role of γδT cells in TNBC. Front Immunol 2024; 15:1420107. [PMID: 38933280 PMCID: PMC11199784 DOI: 10.3389/fimmu.2024.1420107] [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: 04/19/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that presents significant therapeutic challenges due to the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. As a result, conventional hormonal and targeted therapies are largely ineffective, underscoring the urgent need for novel treatment strategies. γδT cells, known for their robust anti-tumor properties, show considerable potential in TNBC treatment as they can identify and eliminate tumor cells without reliance on MHC restrictions. These cells demonstrate extensive proliferation both in vitro and in vivo, and can directly target tumors through cytotoxic effects or indirectly by promoting other immune responses. Studies suggest that expansion and adoptive transfer strategies targeting Vδ2 and Vδ1 γδT cell subtypes have shown promise in preclinical TNBC models. This review compiles and discusses the existing literature on the primary subgroups of γδT cells, their roles in cancer therapy, their contributions to tumor cell cytotoxicity and immune modulation, and proposes potential strategies for future γδT cell-based immunotherapies in TNBC.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xian Zhao
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Chuanxin Ren
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shang Gao
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Qinyu Han
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Min Lu
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xiangqi Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| |
Collapse
|
2
|
Yu H, Yang W, Cao M, Lei Q, Yuan R, Xu H, Cui Y, Chen X, Su X, Zhuo H, Lin L. Mechanism study of ubiquitination in T cell development and autoimmune disease. Front Immunol 2024; 15:1359933. [PMID: 38562929 PMCID: PMC10982411 DOI: 10.3389/fimmu.2024.1359933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
T cells play critical role in multiple immune processes including antigen response, tumor immunity, inflammation, self-tolerance maintenance and autoimmune diseases et. Fetal liver or bone marrow-derived thymus-seeding progenitors (TSPs) settle in thymus and undergo T cell-lineage commitment, proliferation, T cell receptor (TCR) rearrangement, and thymic selections driven by microenvironment composed of thymic epithelial cells (TEC), dendritic cells (DC), macrophage and B cells, thus generating T cells with diverse TCR repertoire immunocompetent but not self-reactive. Additionally, some self-reactive thymocytes give rise to Treg with the help of TEC and DC, serving for immune tolerance. The sequential proliferation, cell fate decision, and selection during T cell development and self-tolerance establishment are tightly regulated to ensure the proper immune response without autoimmune reaction. There are remarkable progresses in understanding of the regulatory mechanisms regarding ubiquitination in T cell development and the establishment of self-tolerance in the past few years, which holds great potential for further therapeutic interventions in immune-related diseases.
Collapse
Affiliation(s)
- Hui Yu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Wenyong Yang
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Min Cao
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Qingqiang Lei
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Renbin Yuan
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - He Xu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yuqian Cui
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xuerui Chen
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xu Su
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Hui Zhuo
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Liangbin Lin
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| |
Collapse
|
3
|
Miccoli A, Pianese V, Bidoli C, Fausto AM, Scapigliati G, Picchietti S. Transcriptome profiling of microdissected cortex and medulla unravels functional regionalization in the European sea bass Dicentrarchus labrax thymus. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109319. [PMID: 38145782 DOI: 10.1016/j.fsi.2023.109319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023]
Abstract
The thymus is a sophisticated primary lymphoid organ in jawed vertebrates, but knowledge on teleost thymus remains scarce. In this study, for the first time in the European sea bass, laser capture microdissection was leveraged to collect two thymic regions based on histological features, namely the cortex and the medulla. The two regions were then processed by RNAseq and in-depth functional transcriptome analyses with the aim of revealing differential gene expression patterns and gene sets enrichments, ultimately unraveling unique microenvironments imperative for the development of functional T cells. The sea bass cortex emerged as a hub of T cell commitment, somatic recombination, chromatin remodeling, cell cycle regulation, and presentation of self antigens from autophagy-, proteasome- or proteases-processed proteins. The cortex therefore accommodated extensive thymocyte proliferation and differentiation up to the checkpoint of positive selection. The medulla instead appeared as the center stage in autoimmune regulation by negative selection and deletion of autoreactive T cells, central tolerance mechanisms and extracellular matrix organization. Region-specific canonical markers of T and non-T lineage cells as well as signals for migration to/from, and trafficking within, the thymus were identified, shedding light on the highly coordinated and exquisitely complex bi-directional interactions among thymocytes and stromal components. Markers ascribable to thymic nurse cells and poorly characterized post-aire mTEC populations were found in the cortex and medulla, respectively. An in-depth data mining also exposed previously un-annotated genomic resources with differential signatures. Overall, our findings contribute to a broader understanding of the relationship between regional organization and function in the European sea bass thymus, and provide essential insights into the molecular mechanisms underlying T-cell mediated adaptive immune responses in teleosts.
Collapse
Affiliation(s)
- A Miccoli
- National Research Council, Institute for Marine Biological Resources and Biotechnology (IRBIM), 60125, Ancona, Italy
| | - V Pianese
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - C Bidoli
- Dept. of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - A M Fausto
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - G Scapigliati
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - S Picchietti
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy.
| |
Collapse
|
4
|
Huang X, Zhang Z, Wang Y, Xu M, Du X, Zhang Y. Circulating miRNAs drive personalized medicine based on subgroup classification in myasthenia gravis patients. Neurol Sci 2023; 44:3877-3884. [PMID: 37402938 DOI: 10.1007/s10072-023-06933-3] [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: 12/25/2022] [Accepted: 06/29/2023] [Indexed: 07/06/2023]
Abstract
Myasthenia gravis (MG) is a classic autoimmune neuromuscular disease with strong clinical heterogeneity. The concept of subgroup classification was proposed to guide the precise treatment of MG. Subgroups based on serum antibodies and clinical features include ocular MG, early-onset MG with AchR antibodies, late-onset MG with AchR antibodies, thymoma-associated MG, MuSK-associated MG, LRP4-associated MG, and seronegative MG. However, reliable objective biomarkers are still needed to reflect the individualized response to therapy. MicroRNAs (miRNAs) are small non-coding RNA molecules which can specifically bind to target genes and regulate gene expression at the post-transcriptional level, and then influence celluar biological processes. MiRNAs play an important role in the pathogenesis of autoimmune diseases, including MG. Several studies on circulating miRNAs in MG have been reported. However, there is rare systematic review to summarize the differences of these miRNAs in different subgroups of MG. Here, we summarize the potential role of circulating miRNAs in different subgroups of MG to promote personalized medicine.
Collapse
Affiliation(s)
- Xiaoyu Huang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhouao Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China
| | - Yingying Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China
| | - Mingming Xu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China
| | - Xue Du
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China
| | - Yong Zhang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, No. 99 Huaihai West Road, Quanshan Distric, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Sertori R, Zhang Y, Wiest DL. Zebrafish: A Tractable Model for Analysis of T Cell Development. Methods Mol Biol 2023; 2580:355-377. [PMID: 36374469 DOI: 10.1007/978-1-0716-2740-2_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
While the zebrafish has for some time been regarded as a powerful model organism with which to study early events in hematopoiesis, recent evidence suggests that it also ideal for unraveling the molecular requirements for T cell development in the thymus. Like mammals, zebrafish possess an adaptive immune system, comprising B lymphocytes as well as both the γδ and αβ lineages of T cells, which develop in the thymus. Moreover, the molecular processes underlying T cell development in zebrafish appear to be remarkably conserved. Thus, findings in the zebrafish model will be of high relevance to the equivalent processes in mammals. Finally, molecular processes can be interrogated in zebrafish far more rapidly than is possible in mammals because the zebrafish possesses many unique advantages. Here, we describe these unique attributes and the methods by which they can be exploited to investigate the role of novel genes in T cell development.
Collapse
Affiliation(s)
- Robert Sertori
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yong Zhang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - David L Wiest
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Kalidasan V, Ravichantar N, Muhd Besari A, Yunus MA, Mohd Yusoff N, Mohamed Z, Theva Das K. Latent HIV-1 provirus in vitro suppression using combinatorial CRISPR/Cas9 strategy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Wang Y, Chang L, Zhu G, Li G, Kong Q, Lv L, Wang Q, Tian C, Li Y, Zhu X, Pan X. Mechanism of thymus rejuvenation in elderly macaques. Rejuvenation Res 2022; 25:223-232. [PMID: 35876435 DOI: 10.1089/rej.2022.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Senile thymus atrophy is an important factor leading to decreased immune function. Repairing the atrophic thymus tissue structure, rebuilding immune function, and replenishing the number of exogenous stem cells may be effective methods. In this study, BMSCs (Bone Marrow Mesenchymal Stem Cells, BMSCs) were intravenously infused into elderly macaques. We found that thymus area was substantially increased, some thymus tissue regeneration was observed, the degree of thymus tissue fibrosis decreased, collagen fibre deposition decreased, cortical and medulla structures emerge gradually, the number of apoptotic cells decreased significantly, inhibit the expression of apoptosis-related proteins. Bone marrow mesenchymal cells inhibit the expression of genes related to aging. By sequencing the mRNA transcriptome in thymus tissue, the expression pattern of the tissue transcriptome tended to be similar to the thymus expression pattern in young macaques compared with the old group, reverse aging-related proteins. Based on the results, an intravenous infusion designed to increase the number of stem cells in the body effectively repairs the structure of the thymus and rebuilds immune function.
Collapse
Affiliation(s)
| | | | - Gaohong Zhu
- The First Affiliated Hospital of Kunming Medical University, Kunming, China;
| | - Gonghua Li
- Kunming Institute of Zoology Chinese Academy of Sciences, Kunming, Yunnan, China;
| | - Qingpeng Kong
- Kunming Institute of Zoology Chinese Academy of Sciences, Kunming, Yunnan, China;
| | - Longbao Lv
- Kunming Institute of Zoology Chinese Academy of Sciences, Kunming, Yunnan, China;
| | | | | | - Ye Li
- 920th hospital, Kunming, China;
| | - Xiangqing Zhu
- 920th hospital , Basic Medical Laboratory, Kunming, China;
| | | |
Collapse
|
8
|
Zapata AG. Lympho-Hematopoietic Microenvironments and Fish Immune System. BIOLOGY 2022; 11:747. [PMID: 35625475 PMCID: PMC9138301 DOI: 10.3390/biology11050747] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022]
Abstract
In the last 50 years information on the fish immune system has increased importantly, particularly that on species of marked commercial interest (i.e., salmonids, cods, catfish, sea breams), that occupy a key position in the vertebrate phylogenetical tree (i.e., Agnatha, Chondrichtyes, lungfish) or represent consolidated experimental models, such as zebrafish or medaka. However, most obtained information was based on genetic sequence analysis with little or no information on the cellular basis of the immune responses. Although jawed fish contain a thymus and lympho-hematopoietic organs equivalents to mammalian bone marrow, few studies have accounted for the presumptive relationships between the organization of these cell microenvironments and the known immune capabilities of the fish immune system. In the current review, we analyze this topic providing information on: (1) The origins of T and B lymphopoiesis in Agnatha and jawed fish; (2) the remarkable organization of the thymus of teleost fish; (3) the occurrence of numerous, apparently unrelated organs housing lympho-hematopoietic progenitors and, presumably, B lymphopoiesis; (4) the existence of fish immunological memory in the absence of germinal centers.
Collapse
Affiliation(s)
- Agustín G. Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; ; Tel.: +34-913-944-979
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
9
|
A cell atlas of microbe-responsive processes in the zebrafish intestine. Cell Rep 2022; 38:110311. [PMID: 35108531 DOI: 10.1016/j.celrep.2022.110311] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 10/28/2021] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Gut microbial products direct growth, differentiation, and development in animal hosts. However, we lack system-wide understanding of cell-specific responses to the microbiome. We profiled cell transcriptomes from the intestine, and associated tissue, of zebrafish larvae raised in the presence or absence of a microbiome. We uncovered extensive cellular heterogeneity in the conventional zebrafish intestinal epithelium, including previously undescribed cell types with known mammalian homologs. By comparing conventional to germ-free profiles, we mapped microbial impacts on transcriptional activity in each cell population. We revealed intricate degrees of cellular specificity in host responses to the microbiome that included regulatory effects on patterning and on metabolic and immune activity. For example, we showed that the absence of microbes hindered pro-angiogenic signals in the developing vasculature, causing impaired intestinal vascularization. Our work provides a high-resolution atlas of intestinal cellular composition in the developing fish gut and details the effects of the microbiome on each cell type.
Collapse
|
10
|
Kernen L, Phan A, Bo J, Herzog EL, Huynh J, Segner H, Baumann L. Estrogens as immunotoxicants: 17α-ethinylestradiol exposure retards thymus development in zebrafish (Danio rerio). AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 242:106025. [PMID: 34837781 DOI: 10.1016/j.aquatox.2021.106025] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 06/13/2023]
Abstract
Estrogenic endocrine disrupting compounds (EEDCs) can cause alterations in sexual development and reproductive function of fish. Growing evidence suggests that EEDCs can also interfere with development and function of innate immunity of fish. The present study examined a potential disruptive effect of EEDCs at field-relevant concentrations on the development of adaptive immunity, more specifically the thymus. Zebrafish (Danio rerio) were exposed from fertilization until 64 days post-fertilization (dpf) to environmentally relevant (3 and 10 ng/L) concentrations of the synthetic estrogen 17α-ethinylestradiol (EE2). The exposure duration covered the period of initial thymus differentiation to maximum growth. Thymus development was assessed by histological and morphometric (thymus area) analysis, thymocyte number, and transcript levels of thymocyte marker genes. Additionally, transcript levels of the estrogen receptors (esr1 and esr2a) were determined. The EE2 exposure altered sexual development (gonad differentiation, transcript levels of hepatic vitellogenin and estrogen receptors) of zebrafish, as expected. At the same time, the EE2 treatment reduced the thymus growth (thymus area, thymocyte number) and transcript levels of thymus marker genes. The expression of the thymic estrogen receptors responded to the EE2 exposure but in a different pattern than the hepatic estrogen receptors. After the 64-day-exposure period, the juvenile fish were transferred into clean water for another 95 days to assess the reversibility of EE2-induced effects. The thymic alterations were found to be reversible in female zebrafish but persisted in males. The present study provides the first evidence that the development of the fish adaptive immune system is sensitive to EEDCs, and that this takes place at concentrations similar to those that disrupt sexual development.
Collapse
Affiliation(s)
- Larissa Kernen
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Audrey Phan
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Jun Bo
- Laboratory of Marine Biology and Ecology, Third Institute of Oceanography, Xiamen 361102, China
| | - Elio L Herzog
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - John Huynh
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland
| | - Lisa Baumann
- Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland; Aquatic Ecology & Toxicology, Centre for Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120 Heidelberg, Germany.
| |
Collapse
|
11
|
Quillien A, Gilbert G, Boulet M, Ethuin S, Waltzer L, Vandel L. Prmt5 promotes vascular morphogenesis independently of its methyltransferase activity. PLoS Genet 2021; 17:e1009641. [PMID: 34153034 PMCID: PMC8248709 DOI: 10.1371/journal.pgen.1009641] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/01/2021] [Accepted: 06/02/2021] [Indexed: 01/02/2023] Open
Abstract
During development, the vertebrate vasculature undergoes major growth and remodeling. While the transcriptional cascade underlying blood vessel formation starts to be better characterized, little is known concerning the role and mode of action of epigenetic enzymes during this process. Here, we explored the role of the Protein Arginine Methyl Transferase Prmt5 in blood vessel formation as well as hematopoiesis using zebrafish as a model system. Through the combination of different prmt5 loss-of-function approaches we highlighted a key role of Prmt5 in both processes. Notably, we showed that Prmt5 promotes vascular morphogenesis through the transcriptional control of ETS transcription factors and adhesion proteins in endothelial cells. Interestingly, using a catalytic dead mutant of Prmt5 and a specific drug inhibitor, we found that while Prmt5 methyltransferase activity was required for blood cell formation, it was dispensable for vessel formation. Analyses of chromatin architecture impact on reporter genes expression and chromatin immunoprecipitation experiments led us to propose that Prmt5 regulates transcription by acting as a scaffold protein that facilitates chromatin looping to promote vascular morphogenesis. Blood vessel formation is an essential developmental process required for the survival of all vertebrates. The vascular anatomy and the mechanisms involved in vessel formation are highly conserved among vertebrates. Hence, we used zebrafish as a model, to decipher the role and the mode of action of Prmt5, an enzyme known to regulate gene expression, in vascular morphogenesis and in blood cell formation in vivo. Using different approaches, we highlighted a key role of Prmt5 during both processes. However, we found that while blood cell formation required Prmt5 enzymatic activity, vascular morphogenesis was independent on its activity. Prmt5 has been proposed as a therapeutic target in many diseases, including cancer. Yet, we show here that Prmt5 acts at least in part independently of its methyltransferase activity to regulate vascular morphogenesis. By shedding light on a mechanism of action of Prmt5 that will be insensitive to enzymatic inhibitors, our data calls forth the design of alternative drugs. In addition, this non-canonical function of Prmt5 may have a more pervasive role than previously thought in physiological conditions, i.e. during development, but also in pathological situations such as in tumor angiogenesis and certainly deserves more attention in the future.
Collapse
Affiliation(s)
- Aurélie Quillien
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- RESTORE, INSERM UMR1301, CNRS UMR5070, Université Paul Sabatier, Université de Toulouse, Toulouse, France
- * E-mail: (AQ); (LV)
| | - Guerric Gilbert
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Manon Boulet
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Séverine Ethuin
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lucas Waltzer
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
| | - Laurence Vandel
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse, France
- Université Clermont Auvergne, CNRS, INSERM, iGReD, Clermont-Ferrand, France
- * E-mail: (AQ); (LV)
| |
Collapse
|
12
|
Hong JH, Kim SH, Kim HG, Jang JH, Son RG, Pack SP, Park YH, Kang P, Jeong KJ, Kim JS, Choi H, Kim SU, Jung YW. Effect of Human or Mouse IL-7 on the Homeostasis of Porcine T Lymphocytes. Immune Netw 2021; 21:e24. [PMID: 34277114 PMCID: PMC8263216 DOI: 10.4110/in.2021.21.e24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/18/2021] [Accepted: 06/20/2021] [Indexed: 12/01/2022] Open
Abstract
Due to the inconsistent fluctuation of blood supply for transfusion, much attention has been paid to the development of artificial blood using other animals. Although mini-pigs are candidate animals, contamination of mini-pig T cells in artificial blood may cause a major safety concern. Therefore, it is important to analyze the cross-reactivity of IL-7, the major survival factor for T lymphocytes, between human, mouse, and mini-pig. Thus, we compared the protein sequences of IL-7 and found that porcine IL-7 was evolutionarily different from human IL-7. We also observed that when porcine T cells were cultured with either human or mouse IL-7, these cells did not increase the survival or proliferation compared to negative controls. These results suggest that porcine T cells do not recognize human or mouse IL-7 as their survival factor.
Collapse
Affiliation(s)
- Ji Hwa Hong
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sang Hoon Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Hyun Gyung Kim
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Jun Ho Jang
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Ryeo Gang Son
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea
| | - Seung Pil Pack
- Department of Biotechnology and Bioinformatics, Korea University, Sejong 30019, Korea
| | - Young-Ho Park
- Futuristic Animal Resource & Research Center (FARRC), Cheongju 28116, Korea
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Philyong Kang
- Futuristic Animal Resource & Research Center (FARRC), Cheongju 28116, Korea
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kang-Jin Jeong
- Futuristic Animal Resource & Research Center (FARRC), Cheongju 28116, Korea
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Ji-Su Kim
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
- Primate Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup 56216, Korea
| | - Hanbyeul Choi
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center (FARRC), Cheongju 28116, Korea
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Yong Woo Jung
- Department of Pharmacy, Korea University, Sejong 30019, Korea
| |
Collapse
|
13
|
Li T, Li R, Zhang T, Zhang H, Song X, Zhai X, Wang J, Xing B, Hou X, Wei L. Identification, cloning, and characterization of Cherry Valley duck CD4 and its antiviral immune responses. Poult Sci 2021; 100:101262. [PMID: 34273645 PMCID: PMC8287243 DOI: 10.1016/j.psj.2021.101262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/11/2021] [Accepted: 05/12/2021] [Indexed: 11/19/2022] Open
Abstract
CD4 protein is a single chain transmembrane glycoprotein and has a broad functionality beyond cell-mediated immunity. In this study, we cloned the full-length coding sequence (CDS) of duck CD4 (duCD4) and analyzed its sequence and structure, and expression levels in several tissues. It consists of 1,449 nucleotides and encodes a 482 amino acid protein. The putative protein of duCD4 consisted of an N-terminal signal peptide, three immunoglobulins and one immunoglobulins-like domain in its central, one terminal transmembrane region, and a C-terminal domain of the CD4 T cell receptor. The duCD4 also has the typical signature “CXC” of CD4s. The multiple sequence alignment suggests duCD4 has four potential N-glycosylation sites and the phylogenetic analysis suggests duCD4 shares greater similarity with avian than other vertebrates. Quantitative real-time PCR analysis showed that duCD4 mRNA transcripts are widely distributed in the healthy Cherry Valley duck, and the highest level in the thymus. During the virus infection, the obvious change of duCD4 expression was observed in the spleen, lung and brain, which suggesting that duCD4 could be involved in the host's immune response to multiple types of viruses. Our research studied the characterization, tissue distribution, and antiviral immune responses of duCD4.
Collapse
Affiliation(s)
- Tianxu Li
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Rong Li
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province 266109, China
| | - Tingting Zhang
- Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, College of Basic Medical Sciences, Shandong First Medical University, Tai'an City, Shandong Province 271000, China
| | - Huihui Zhang
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xingdong Song
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xinyu Zhai
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Jinchao Wang
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Bin Xing
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Xiaolan Hou
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China
| | - Liangmeng Wei
- Sino-German Cooperative Research Centre for Zoonosis of Animal Origin of Shandong Province, Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province 271018, China; Collaborative Innovation Center for the Origin and Control of Emerging Infectious Diseases, College of Basic Medical Sciences, Shandong First Medical University, Tai'an City, Shandong Province 271000, China.
| |
Collapse
|
14
|
Loontiens S, Dolens AC, Strubbe S, Van de Walle I, Moore FE, Depestel L, Vanhauwaert S, Matthijssens F, Langenau DM, Speleman F, Van Vlierberghe P, Durinck K, Taghon T. PHF6 Expression Levels Impact Human Hematopoietic Stem Cell Differentiation. Front Cell Dev Biol 2020; 8:599472. [PMID: 33251223 PMCID: PMC7672048 DOI: 10.3389/fcell.2020.599472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/15/2020] [Indexed: 01/10/2023] Open
Abstract
Transcriptional control of hematopoiesis involves complex regulatory networks and functional perturbations in one of these components often results in malignancies. Loss-of-function mutations in PHF6, encoding a presumed epigenetic regulator, have been primarily described in T cell acute lymphoblastic leukemia (T-ALL) and the first insights into its function in normal hematopoiesis only recently emerged from mouse modeling experiments. Here, we investigated the role of PHF6 in human blood cell development by performing knockdown studies in cord blood and thymus-derived hematopoietic precursors to evaluate the impact on lineage differentiation in well-established in vitro models. Our findings reveal that PHF6 levels differentially impact the differentiation of human hematopoietic progenitor cells into various blood cell lineages, with prominent effects on lymphoid and erythroid differentiation. We show that loss of PHF6 results in accelerated human T cell development through reduced expression of NOTCH1 and its downstream target genes. This functional interaction in developing thymocytes was confirmed in vivo using a phf6-deficient zebrafish model that also displayed accelerated developmental kinetics upon reduced phf6 or notch1 activation. In summary, our work reveals that appropriate control of PHF6 expression is important for normal human hematopoiesis and provides clues towards the role of PHF6 in T-ALL development.
Collapse
Affiliation(s)
- Siebe Loontiens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | - Steven Strubbe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | | | - Finola E. Moore
- Molecular Pathology and Cancer Center, Massachusetts General Hospital, Boston, MA, United States
| | - Lisa Depestel
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Suzanne Vanhauwaert
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Filip Matthijssens
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - David M. Langenau
- Molecular Pathology and Cancer Center, Massachusetts General Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Frank Speleman
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Pieter Van Vlierberghe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Kaat Durinck
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
15
|
Ekong E, Ndembi N, Okonkwo P, Dakum P, Idoko J, Banigbe B, Okuma J, Agaba P, Blattner W, Adebamowo C, Charurat M. Epidemiologic and viral predictors of antiretroviral drug resistance among persons living with HIV in a large treatment program in Nigeria. AIDS Res Ther 2020; 17:7. [PMID: 32066473 PMCID: PMC7027291 DOI: 10.1186/s12981-020-0261-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 01/23/2020] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Expanded access to combination antiretroviral therapy (cART) throughout sub-Saharan Africa over the last decade has remarkably improved the prognosis of persons living with HIV (PLWH). However, some PLWH experience virologic rebound after a period of viral suppression, usually followed by selection of drug resistant virus. Determining factors associated with drug resistance can inform patient management and healthcare policies, particularly in resource-limited settings where drug resistance testing is not routine. METHODS A case-control study was conducted using data captured from an electronic medical record in a large treatment program in Nigeria. Cases PLWH receiving cART who developed acquired drug resistance (ADR) and controls were those without ADR between 2004 and 2011. Each case was matched to up to 2 controls by sex, age, and education. Logistic regression was used estimate odds ratios (ORs) and 95% confidence intervals (CIs) for factors associated with ADR. RESULTS We evaluated 159 cases with ADR and 299 controls without ADR. In a multivariate model, factors associated with ADR included older age (OR = 2.35 [age 30-40 years 95% CI 1.29, 4.27], age 41 + years OR = 2.31 [95% CI 1.11, 4.84], compared to age 17-30), higher education level (secondary OR 2.14 [95% CI 1.1.11-4.13]), compared to primary and tertiary), non-adherence to care (OR = 2.48 [95% CI 1.50-4.00]), longer treatment duration (OR = 1.80 [95% CI 1.37-2.35]), lower CD4 count((OR = 0.95 [95% CI 0.95-0.97]) and higher viral load (OR = 1.97 [95% CI 1.44-2.54]). CONCLUSIONS Understanding these predictors may guide programs in developing interventions to identify patients at risk of developing ADR and implementing prevention strategies.
Collapse
Affiliation(s)
- Ernest Ekong
- Department of Prevention, Care and Treatment, Institute of Human Virology-Nigeria (IHVN), Federal Capital Territory, Plot 252, Herbert Macaulay Way, Central Business District, Abuja, Nigeria.
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Nicaise Ndembi
- Department of Prevention, Care and Treatment, Institute of Human Virology-Nigeria (IHVN), Federal Capital Territory, Plot 252, Herbert Macaulay Way, Central Business District, Abuja, Nigeria.
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | - Patrick Dakum
- Department of Prevention, Care and Treatment, Institute of Human Virology-Nigeria (IHVN), Federal Capital Territory, Plot 252, Herbert Macaulay Way, Central Business District, Abuja, Nigeria
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - John Idoko
- Jos University Teaching Hospital, Jos, Nigeria
| | | | - James Okuma
- Department of Prevention, Care and Treatment, Institute of Human Virology-Nigeria (IHVN), Federal Capital Territory, Plot 252, Herbert Macaulay Way, Central Business District, Abuja, Nigeria
| | | | - William Blattner
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Clement Adebamowo
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Manhattan Charurat
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Li Y, Lei X, Lu H, Guo W, Wu S, Yin Z, Sun Q, Yang X. Age-Related Changes on CD40 Promotor Methylation and Immune Gene Expressions in Thymus of Chicken. Front Immunol 2018; 9:2731. [PMID: 30519246 PMCID: PMC6259354 DOI: 10.3389/fimmu.2018.02731] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 11/06/2018] [Indexed: 12/29/2022] Open
Abstract
One hundred and twenty one-day-old breeder cocks, included 15 cages of 8 birds each, were fed to learn the aging's effect on chicken's thymus immunity. At 2 (2-W) and 40 (40-W) weeks of age, one chicken each cage was randomly chosen and slaughtered to get the thymus sample. The results showed that thymus weight and morphology of 40-W group were far different from that of 2-W group, and exhibited a property of degeneration. Considering this phenotype variance, we analyzed the thymus' transcriptome to investigate the molecular mechanism that had been implicated in this phenotype diversity with age. Pearson correlation coefficients and principal component analysis indicated that two major populations corresponding to 40-W and 2-W group were identified, and 1949 differentially expressed genes (DEGs, 1722 up-regulated and 127 down-regulated) were obtained. Results of GO and KEGG pathway enrichment found that 4 significantly enriched KEGG pathways (Cytokine-cytokine receptor interaction, Intestinal immune network for IgA production, Toll-like receptor signaling pathway, AGE-RAGE signaling pathway in diabetic complications) related to immunoregulation were screened between 40-W and 2-W group. These results confirmed that thymus immunity of chickens had a strong age-related correlation. DEGs related to these 4 enriched KEGG pathways were suppressed in the thymus of 2-W group, this indicated that thymus immunity of 2-weeks-age chick was down-regulated. CD40 is involved in 3 of the 4 significantly enriched pathways, and it is critical for thymus immune-regulation. CD40 promoter methylation level of 2-W group was higher than that of 40-W group, it is consistent with the transcriptional differences of the gene. Our study concluded that thymus immunity of chicken was varied with age. Compared to the 40-W group, thymus immunity of 2-W group was down-regulated, and in a status of hypo-activation on the whole, and these effects might be related to CD40 suppression induced by promoter hyper-methylation of the gene.
Collapse
Affiliation(s)
- Yulong Li
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Xinyu Lei
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Hong Lu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Wei Guo
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Zhenchen Yin
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Qingzhu Sun
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Xi'an, China
| |
Collapse
|
17
|
Wei C, Guo D, Li Y, Zhang K, Liang G, Li Y, Ma Y, Liu J, Li Y. Profiling analysis of 17β-estradiol-regulated lncRNAs in mouse thymic epithelial cells. Physiol Genomics 2018; 50:553-562. [DOI: 10.1152/physiolgenomics.00098.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Thymus is the primary organ for T cell differentiation and maturation. Many studies have demonstrated that estrogen plays a crucial role in thymic epithelial cell (TEC) proliferation during thymic involution. LncRNAs are involved in various biological processes; however, estrogen-mediated lncRNA expression in TECs has not been yet reported. To address this question, the mouse medullary thymic epithelial cell line 1 (MTEC1) was treated with 17β-estradiol (E2). By using CCK8 assay and flow cytometry, we found that E2 was able to inhibit viability and proliferation of MTEC1 cells. The expression profiles of lncRNAs in MTEC1 cells with or without E2 treatment were then measured by RNA-Seq, and a total of 962 lncRNAs and 2,469 mRNAs were shown to be differentially expressed. The reliability of RNA-Seq was confirmed by quantitative RT-PCR. Correlation analysis was conducted to investigate the potential function of lncRNAs. According to gene ontology (GO) analysis, differentially expressed lncRNAs were mainly related to cell proliferation, cell cycle and cell apoptosis. KEGG pathway analysis indicated that these lncRNAs were associated with several pathways, namely immunological activity, metabolism and cytokine-cytokine receptor interaction. In conclusion, our study provided a novel direction for studying the relationship between lncRNAs and E2 in the thymus.
Collapse
Affiliation(s)
- Chaonan Wei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Dongguang Guo
- School of Life Science and Technology, Xinxiang University, Xinxiang, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Kaizhao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Guan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jilong Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Zhang K, Tan X, Li Y, Liang G, Ning Z, Ma Y, Li Y. Transcriptional profiling analysis of Zearalenone-induced inhibition proliferation on mouse thymic epithelial cell line 1. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 153:135-141. [PMID: 29425844 DOI: 10.1016/j.ecoenv.2018.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/28/2017] [Accepted: 01/03/2018] [Indexed: 06/08/2023]
Abstract
Zearalenone (ZEA) was a mycotoxin biosynthesized by a variety of Fusarium fungi via a polypeptide pathway. ZEA has significant toxic reaction on immune cells. Thymic epithelial cells (TECs) as a crucial constituent of thymic stroma can provide unique microenvironment for thymocyte maturation, but the mechanism of ZEA affecting the TECs is poorly understood. The basic data about gene expression differences for the ZEA on thymic epithelial cell line 1 (MTEC1) will help us to elucidate this mechanism. Here, cell viability and proliferation assay and transcriptome sequencing on MTEC1 treated with ZEA were performed. 4188 differentially expressed genes (DEGs) between ZEA treated and control groups were identified, confirmed and analyzed. Our results showed that 10-50μg/ml ZEA significantly inhibited MTEC1 proliferation and arrested cell cycle at G2/M phase. Gene ontology and KEGG pathway analysis revealed that Chemokine, JAK-STAT and Toll-like receptor signaling pathway, were involved in the cell cycle pathway. 16 key genes involved in the cell cycle processes were validated and the results suggested that Mitotic catastrophe (MC) may take part in ZEA inhibition of METC1 cell proliferation. These data highlighted the importance of cell cycle pathway in MTEC1 treated with ZEA, and will contribute to get the molecular mechanisms of ZEA inhibition of MTEC1 cell proliferation.
Collapse
Affiliation(s)
- Kaizhao Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiaotong Tan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Guan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
19
|
Liu ZZ, Wang ZL, Choi TI, Huang WT, Wang HT, Han YY, Zhu LY, Kim HT, Choi JH, Lee JS, Kim HG, Zhao J, Chen Y, Lu Z, Tian XL, Pan BX, Li BM, Kim CH, Xu HA. Chd7 Is Critical for Early T-Cell Development and Thymus Organogenesis in Zebrafish. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1043-1058. [PMID: 29353058 DOI: 10.1016/j.ajpath.2017.12.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 12/31/2022]
Abstract
Coloboma, heart defect, atresia choanae, retarded growth and development, genital hypoplasia, ear anomalies/deafness (CHARGE) syndrome is a congenital disorder affecting multiple organs and mainly caused by mutations in CHD7, a gene encoding a chromatin-remodeling protein. Immunodeficiency and reduced T cells have been noted in CHARGE syndrome. However, the mechanisms underlying T lymphopenia are largely unexplored. Herein, we observed dramatic decrease of T cells in both chd7knockdown and knockout zebrafish embryos. Unexpectedly, hematopoietic stem and progenitor cells and, particularly, lymphoid progenitor cells were increased peripherally in nonthymic areas in chd7-deficient embryos, unlikely to contribute to the T-cell decrease. Further analysis demonstrated that both the organogenesis and homing function of the thymus were seriously impaired. Chd7 might regulate thymus organogenesis through modulating the development of both neural crest cell-derived mesenchyme and pharyngeal endoderm-derived thymic epithelial cells. The expression of foxn1, a central regulator of thymic epithelium, was remarkably down-regulated in the pharyngeal region in chd7-deficient embryos. Moreover, the T-cell reduction in chd7-deficient embryos was partially rescued by overexpressing foxn1, suggesting that restoring thymic epithelium may be a potential therapeutic strategy for treating immunodeficiency in CHARGE syndrome. Collectively, the results indicated that chd7 was critical for thymic development and T-lymphopenia in CHARGE syndrome may be mainly attributed to the defects of thymic organogenesis. The current finding may benefit the diagnosis and therapy of T lymphopenia and immunodeficiency in CHARGE syndrome.
Collapse
Affiliation(s)
- Zhi-Zhi Liu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Zi-Long Wang
- Institute of Life Science, Nanchang University, Nanchang, China; Queen Mary School, Nanchang University, Nanchang, China
| | - Tae-Ik Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Wen-Ting Huang
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Han-Tsing Wang
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Ying-Ying Han
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Lou-Yin Zhu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Hyun-Taek Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jung-Hwa Choi
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jin-Soo Lee
- National Cancer Center, Goyang, Republic of Korea
| | - Hyung-Goo Kim
- Department of Obstetrics and Gynecology, Augusta University, Augusta, Georgia; Children's Hospital of Jiang Xi, Nanchang, China; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia
| | - Jian Zhao
- Hospital of Nanchang University, Nanchang University, Nanchang, China
| | - Yue Chen
- Children's Hospital of Jiang Xi, Nanchang, China
| | - Zhuo Lu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China
| | - Xiao-Li Tian
- School of Life Sciences, Nanchang University, Nanchang, China
| | - Bing-Xing Pan
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Bao-Ming Li
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, Republic of Korea.
| | - Hong A Xu
- Institute of Life Science, Nanchang University, Nanchang, China; School of Life Sciences, Nanchang University, Nanchang, China; Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China.
| |
Collapse
|
20
|
Bieńkowska A, Kiernozek E, Kozlowska E, Bugajski Ł, Drela N. A new approach to the role of IL-7 and TGF-ß in the in vitro generation of thymus-derived CD4+CD25+Foxp3+ regulatory T cells. Cytokine 2017; 102:107-116. [PMID: 28826649 DOI: 10.1016/j.cyto.2017.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/28/2017] [Accepted: 07/24/2017] [Indexed: 12/28/2022]
Abstract
Thymus-derived regulatory T cells of CD4+CD25+Foxp3+ phenotype develop as a functional, mature population playing an essential role in self-tolerance and immune homeostasis, and exhibiting therapeutic potential to inhibit adverse immune response. Despite intensive research on thymus-derived Tregs, the knowledge about agents involved in their generation, survival, proliferation, and biological functions is still insufficient. In this research we have focused on the role of selected cytokines in previously developed in vitro model based on the application of anti-CD3 monoclonal antibodies. We have demonstrated an essential role of IL-7 and TGF-β in the generation of thymus-derived Tregs in the co-culture of thymocytes and JAWS II cells. In addition, in vitro generated Tregs exhibited their suppressive function similarly to Tregs sorted from freshly isolated thymus.
Collapse
Affiliation(s)
- Anna Bieńkowska
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Ewelina Kiernozek
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Ewa Kozlowska
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Łukasz Bugajski
- The Laboratory of Cytometry, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Nadzieja Drela
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
21
|
Pillay LM, Mackowetzky KJ, Widen SA, Waskiewicz AJ. Somite-Derived Retinoic Acid Regulates Zebrafish Hematopoietic Stem Cell Formation. PLoS One 2016; 11:e0166040. [PMID: 27861498 PMCID: PMC5115706 DOI: 10.1371/journal.pone.0166040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 10/11/2016] [Indexed: 01/14/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are multipotent progenitors that generate all vertebrate adult blood lineages. Recent analyses have highlighted the importance of somite-derived signaling factors in regulating HSC specification and emergence from dorsal aorta hemogenic endothelium. However, these factors remain largely uncharacterized. We provide evidence that the vitamin A derivative retinoic acid (RA) functions as an essential regulator of zebrafish HSC formation. Temporal analyses indicate that RA is required for HSC gene expression prior to dorsal aorta formation, at a time when the predominant RA synthesis enzyme, aldh1a2, is strongly expressed within the paraxial mesoderm and somites. Previous research implicated the Cxcl12 chemokine and Notch signaling pathways in HSC formation. Consequently, to understand how RA regulates HSC gene expression, we surveyed the expression of components of these pathways in RA-depleted zebrafish embryos. During somitogenesis, RA-depleted embryos exhibit altered expression of jam1a and jam2a, which potentiate Notch signaling within nascent endothelial cells. RA-depleted embryos also exhibit a severe reduction in the expression of cxcr4a, the predominant Cxcl12b receptor. Furthermore, pharmacological inhibitors of RA synthesis and Cxcr4 signaling act in concert to reduce HSC formation. Our analyses demonstrate that somite-derived RA functions to regulate components of the Notch and Cxcl12 chemokine signaling pathways during HSC formation.
Collapse
Affiliation(s)
- Laura M Pillay
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Kacey J Mackowetzky
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Sonya A Widen
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| | - Andrew Jan Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada.,Women & Children's Health Research Institute, University of Alberta, Edmonton, Alberta, T6G 2E9, Canada
| |
Collapse
|
22
|
Pezzi N, Assis AF, Cotrim-Sousa LC, Lopes GS, Mosella MS, Lima DS, Bombonato-Prado KF, Passos GA. Aire knockdown in medullary thymic epithelial cells affects Aire protein, deregulates cell adhesion genes and decreases thymocyte interaction. Mol Immunol 2016; 77:157-73. [PMID: 27505711 DOI: 10.1016/j.molimm.2016.08.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 07/12/2016] [Accepted: 08/02/2016] [Indexed: 12/15/2022]
Abstract
We demonstrate that even a partial reduction of Aire mRNA levels by siRNA-induced Aire knockdown (Aire KD) has important consequences to medullary thymic epithelial cells (mTECs). Aire knockdown is sufficient to reduce Aire protein levels, impair its nuclear location, and cause an imbalance in large-scale gene expression, including genes that encode cell adhesion molecules. These genes drew our attention because adhesion molecules are implicated in the process of mTEC-thymocyte adhesion, which is critical for T cell development and the establishment of central self-tolerance. Accordingly, we consider the following: 1) mTECs contribute to the elimination of self-reactive thymocytes through adhesion; 2) Adhesion molecules play a crucial role during physical contact between these cells; and 3) Aire is an important transcriptional regulator in mTECs. However, its role in controlling mTEC-thymocyte adhesion remains unclear. Because Aire controls adhesion molecule genes, we hypothesized that the disruption of its expression could influence mTEC-thymocyte interaction. To test this hypothesis, we used a murine Aire(+) mTEC cell line as a model system to reproduce mTEC-thymocyte adhesion in vitro. Transcriptome analysis of the mTEC cell line revealed that Aire KD led to the down-modulation of more than 800 genes, including those encoding for proteins involved in cell adhesion, i.e., the extracellular matrix constituent Lama1, the CAM family adhesion molecules Vcam1 and Icam4, and those that encode peripheral tissue antigens. Thymocytes co-cultured with Aire KD mTECs had a significantly reduced capacity to adhere to these cells. This finding is the first direct evidence that Aire also plays a role in controlling mTEC-thymocyte adhesion.
Collapse
Affiliation(s)
- Nicole Pezzi
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Amanda Freire Assis
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Larissa Cotrim Cotrim-Sousa
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Gabriel Sarti Lopes
- Department of Cellular and Molecular Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, SP, Brazil
| | - Maritza Salas Mosella
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Djalma Sousa Lima
- Department of Cellular and Molecular Biology, Ribeirão Preto Medical School, USP, Ribeirão Preto, SP, Brazil
| | - Karina F Bombonato-Prado
- Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil; Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
23
|
Abstract
While zebrafish have for some time been regarded as a powerful model organism with which to study early events in hematopoiesis, recent evidence suggests that it also ideal for unraveling the molecular requirements for T cell development in the thymus. Like mammals, zebrafish possess an adaptive immune system, comprising B lymphocytes as well as both the γδ and αβ lineages of T cells, which develop in the thymus. Moreover, the molecular processes underlying T cell development in zebrafish appear to be remarkably conserved. Thus, findings in the zebrafish model will be of high relevance to the equivalent processes in mammals. Finally, molecular processes can be interrogated in zebrafish far more rapidly than is possible in mammals because the zebrafish possesses many unique advantages. These unique attributes, and the methods by which they can be exploited to investigate the role of novel genes in T cell development, are described here.
Collapse
Affiliation(s)
- Yong Zhang
- Blood Cell Development and Function Program, Fox Chase Cancer Center, 333 Cottman Avenue, R364, Reimann Building, Philadelphia, PA, 19111, USA,
| | | |
Collapse
|
24
|
Comprehensive Survey of miRNA-mRNA Interactions Reveals That Ccr7 and Cd247 (CD3 zeta) are Posttranscriptionally Controlled in Pancreas Infiltrating T Lymphocytes of Non-Obese Diabetic (NOD) Mice. PLoS One 2015; 10:e0142688. [PMID: 26606254 PMCID: PMC4659659 DOI: 10.1371/journal.pone.0142688] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/26/2015] [Indexed: 01/14/2023] Open
Abstract
In autoimmune type 1 diabetes mellitus (T1D), auto-reactive clones of CD4+ and CD8+ T lymphocytes in the periphery evolve into pancreas-infiltrating T lymphocytes (PILs), which destroy insulin-producing beta-cells through inflammatory insulitis. Previously, we demonstrated that, during the development of T1D in non-obese diabetic (NOD) mice, a set of immune/inflammatory reactivity genes were differentially expressed in T lymphocytes. However, the posttranscriptional control involving miRNA interactions that occur during the evolution of thymocytes into PILs remains unknown. In this study, we postulated that miRNAs are differentially expressed during this period and that these miRNAs can interact with mRNAs involved in auto-reactivity during the progression of insulitis. To test this hypothesis, we used NOD mice to perform, for the first time, a comprehensive survey of miRNA and mRNA expression as thymocytes mature into peripheral CD3+ T lymphocytes and, subsequently, into PILs. Reconstruction of miRNA-mRNA interaction networks for target prediction revealed the participation of a large set of miRNAs that regulate mRNA targets related to apoptosis, cell adhesion, cellular regulation, cellular component organization, cellular processes, development and the immune system, among others. The interactions between miR-202-3p and the Ccr7 chemokine receptor mRNA or Cd247 (Cd3 zeta chain) mRNA found in PILs are highlighted because these interactions can contribute to a better understanding of how the lack of immune homeostasis and the emergence of autoimmunity (e.g., T1D) can be associated with the decreased activity of Ccr7 or Cd247, as previously observed in NOD mice. We demonstrate that these mRNAs are controlled at the posttranscriptional level in PILs.
Collapse
|
25
|
Mendoza L, Méndez A. A dynamical model of the regulatory network controlling lymphopoiesis. Biosystems 2015; 137:26-33. [PMID: 26408858 DOI: 10.1016/j.biosystems.2015.09.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/22/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022]
Abstract
Due to the large number of diseases associated to a malfunction of the hematopoietic system, there is an interest in knowing the molecular mechanisms controlling the differentiation of blood cell lineages. However, the structure and dynamical properties of the underlying regulatory network controlling this process is not well understood. This manuscript presents a regulatory network of 81 nodes, representing several types of molecules that regulate each other during the process of lymphopoiesis. The regulatory interactions were inferred mostly from published experimental data. However, 15 out of 159 regulatory interactions are predictions arising from the present study. The network is modelled as a continuous dynamical system, in the form of a set of differential equations. The dynamical behaviour of the model describes the differentiation process from the common lymphocyte precursor (CLP) to several mature B and T cell types; namely, plasma cell (PC), cytotoxic T lymphocyte (CTL), T helper 1 (Th1), Th2, Th17, and T regulatory (Treg) cells. The model qualitatively recapitulates key cellular differentiation events, being able to represent the directional and branched nature of lymphopoiesis, going from a multipotent progenitor to fully differentiated cell types.
Collapse
Affiliation(s)
- Luis Mendoza
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico.
| | - Akram Méndez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, Mexico; Programa de Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, México, Mexico
| |
Collapse
|
26
|
Inghirami G, Chan WC, Pileri S. Peripheral T-cell and NK cell lymphoproliferative disorders: cell of origin, clinical and pathological implications. Immunol Rev 2015; 263:124-59. [PMID: 25510275 DOI: 10.1111/imr.12248] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell lymphoproliferative disorders are a heterogeneous group of neoplasms with distinct clinical-biological properties. The normal cellular counterpart of these processes has been postulated based on functional and immunophenotypic analyses. However, T lymphocytes have been proven to be remarkably capable of modulating their properties, adapting their function in relationship with multiple stimuli and to the microenvironment. This impressive plasticity is determined by the equilibrium among a pool of transcription factors and by DNA chromatin regulators. It is now proven that the acquisition of specific genomic defects leads to the enforcement/activation of distinct pathways, which ultimately alter the preferential activation of defined regulators, forcing the neoplastic cells to acquire features and phenotypes distant from their original fate. Thus, dissecting the landscape of the genetic defects and their functional consequences in T-cell neoplasms is critical not only to pinpoint the origin of these tumors but also to define innovative mechanisms to re-adjust an unbalanced state to which the tumor cells have become addicted and make them vulnerable to therapies and targetable by the immune system. In our review, we briefly describe the pathological and clinical aspects of the T-cell lymphoma subtypes as well as NK-cell lymphomas and then focus on the current understanding of their pathogenesis and the implications on diagnosis and treatment.
Collapse
Affiliation(s)
- Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy; Department of Pathology, and NYU Cancer Center, New York University School of Medicine, New York, NY, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | | | | |
Collapse
|
27
|
Passos GA, Mendes-da-Cruz DA, Oliveira EH. The Thymic Orchestration Involving Aire, miRNAs, and Cell-Cell Interactions during the Induction of Central Tolerance. Front Immunol 2015; 6:352. [PMID: 26236310 PMCID: PMC4500981 DOI: 10.3389/fimmu.2015.00352] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 06/29/2015] [Indexed: 01/23/2023] Open
Abstract
Developing thymocytes interact sequentially with two distinct structures within the thymus: the cortex and medulla. Surviving single-positive and double-positive thymocytes from the cortex migrate into the medulla, where they interact with medullary thymic epithelial cells (mTECs). These cells ectopically express a vast set of peripheral tissue antigens (PTAs), a property termed promiscuous gene expression that is associated with the presentation of PTAs by mTECs to thymocytes. Thymocyte clones that have a high affinity for PTAs are eliminated by apoptosis in a process termed negative selection, which is essential for tolerance induction. The Aire gene is an important factor that controls the expression of a large set of PTAs. In addition to PTAs, Aire also controls the expression of miRNAs in mTECs. These miRNAs are important in the organization of the thymic architecture and act as posttranscriptional controllers of PTAs. Herein, we discuss recent discoveries and highlight open questions regarding the migration and interaction of developing thymocytes with thymic stroma, the ectopic expression of PTAs by mTECs, the association between Aire and miRNAs and its effects on central tolerance.
Collapse
Affiliation(s)
- Geraldo Aleixo Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, São Paulo , Brazil ; Disciplines of Genetics and Molecular Biology, Department of Morphology, Physiology and Basic Pathology, School of Dentistry of Ribeirão Preto, University of São Paulo , Ribeirão Preto, São Paulo , Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation , Rio de Janeiro, Rio de Janeiro , Brazil
| | - Ernna Hérida Oliveira
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo , Ribeirão Preto, São Paulo , Brazil
| |
Collapse
|
28
|
Vasil’ev KA, Polevshchikov AV. Thymus development in early ontogeny: A comparative aspect. Russ J Dev Biol 2015. [DOI: 10.1134/s106236041503008x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Leyva-Rangel JP, de Los Angeles Hernández-Cueto M, Galan-Enriquez CS, López-Medina M, Ortiz-Navarrete V. Bacterial clearance reverses a skewed T-cell repertoire induced by Salmonella infection. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:209-23. [PMID: 26417438 PMCID: PMC4578521 DOI: 10.1002/iid3.60] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 11/06/2022]
Abstract
Salmonella typhimurium invades the spleen, liver, and peripheral lymph nodes and has recently been detected in the bone marrow and thymus, resulting in a reduced thymic size and a decline in the total number of thymic cells. A specific deletion of the double-positive cell subset has been characterized, yet the export of mature T cells to the periphery remains normal. We analyzed Salmonella pathogenesis regarding thymic structure and the T-cell maturation process. We demonstrate that, despite alterations in the thymic structure, T-cell development is maintained during Salmonella infection, allowing the selection of single-positive T-cell clones expressing particular T-cell receptor beta chains (TCR-Vβ). Moreover, the treatment of infected mice with an antibiotic restored the normal thymic architecture and thymocyte subset distribution. Additionally, the frequency of TCR-Vβ usage after treatment was comparable to that in non-infected mice. However, bacteria were still recovered from the thymus after 1 month of treatment. Our data reveal that a skewed T-cell developmental process is present in the Salmonella-infected thymus that alters the TCR-Vβ usage frequency. Likewise, the post-treatment persistence of Salmonella reveals a novel function of the thymus as a potential reservoir for this infectious agent.
Collapse
Affiliation(s)
- Jessica P Leyva-Rangel
- Doctorado en Ciencias Biomédicas Facultad de Medicina, UNAM Mexico City, CP 045510, Mexico ; Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | | | - Carlos-Samuel Galan-Enriquez
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | - Marcela López-Medina
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politecnico Nacional (CINVESTAV) Mexico City
| |
Collapse
|
30
|
Gan Z, Wang B, Zhou W, Lu Y, Zhu W, Tang J, Jian J, Wu Z. Molecular and functional characterization of CD59 from Nile tilapia (Oreochromis niloticus) involved in the immune response to Streptococcus agalactiae. FISH & SHELLFISH IMMUNOLOGY 2015; 44:50-59. [PMID: 25661843 DOI: 10.1016/j.fsi.2015.01.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/22/2015] [Accepted: 01/26/2015] [Indexed: 06/04/2023]
Abstract
CD59, the major inhibitor of membrane attack complex, plays a crucial role in regulation of complement activation. In this paper, a CD59 gene of Nile tilapia, Oreochromis niloticus (designated as On-CD59) was cloned and its expression pattern under the stimulation of Streptococcus agalactiae was investigated. Sequence analysis showed main structural features required for complement-inhibitory activity were detected in the deduced amino acid sequence of On-CD59. In healthy Nile tilapia, the On-CD59 transcripts could be detected in all the examined tissues, with the most abundant expression in the brain. When immunized with inactivated S. agalactiae, there was a clear time-dependent expression pattern of On-CD59 in the skin, brain, head kidney, thymus and spleen, with quite different kinetic expressions. The assays for the complement-inhibitory activity suggested that recombinant On-CD59 protein had a species-selective inhibition of complement. Moreover, our works showed that recombinant On-CD59 protein may possess both binding activities to PGN and LTA and inhibiting activity of S. agalactiae. These findings indicated that On-CD59 may play important roles in the immune response to S. agalactiae in Nile tilapia.
Collapse
Affiliation(s)
- Zhen Gan
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Bei Wang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Wei Zhou
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Yishan Lu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China.
| | - Weiwei Zhu
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Jufen Tang
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - JiChang Jian
- College of Fishery, Guangdong Ocean University, Zhanjiang, 524025, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| | - Zaohe Wu
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, 524025, China; Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Insititutes, Zhanjiang, 524025, China
| |
Collapse
|
31
|
Liu Y, He S, Zhang Y, Xia W, Li M, Zhang C, Gao F. Effects of Intrauterine Growth Restriction During Late Pregnancy on the Development of the Ovine Fetal Thymus and the T-Lymphocyte Subpopulation. Am J Reprod Immunol 2015; 74:26-37. [PMID: 25708257 DOI: 10.1111/aji.12371] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/28/2015] [Indexed: 12/21/2022] Open
Abstract
PROBLEM The retarded development of fetal thymus in intrauterine growth restriction (IUGR) from maternal undernutrition during late pregnancy destroys the tridimensional structure and modifies the development of fetal T lymphocytes. The mechanisms, however, remain unclear. The objective of this study was to investigate the effect of IUGR during late pregnancy on the development of the ovine fetal thymus and the T-lymphocyte subpopulation. METHOD OF STUDY Eighteen time-mated ewes with singleton fetuses were allocated to three groups at day 90 of pregnancy: restricted group 1 (RG1, 0.18 MJ ME/BW(0.75) /day, n = 6), restricted group 2 (RG2, 0.33 MJ ME/BW(0.75) /day, n = 6) and a control group (CG, ad libitum, 0.67 MJ ME/BW(0.75) /day, n = 6). Fetuses were recovered at slaughter on day 140. RESULTS Fetuses in RG1 exhibited decreased (P < 0.05) thymic weight, cortical thickness, cortical:medullary, DNA content, total antioxidant capacity, and superoxide dismutase; intermediate changes were found in RG2 fetuses, including decreased thymic weight, cortical thickness, and DNA content (P < 0.05). The reductions (P < 0.05) of CD4(+) CD8(+) T cells, relative mRNA expression of keratin 8, recombination activating gene 1 (RAG1), and B-cell lymphoma 2 (Bcl-2) were found in both restricted groups. In addition, there was reduced mRNA expression (P < 0.05) of T-cell receptor, apoptosis antigen 1 ligand, and RAG2 in the RG1 group. In contrast, increases in glutathione peroxidase, malondialdehyde, caspase-3, Cytochrome c, and CD4(+) T cells were observed (P < 0.05), and higher mRNA expressions (P < 0.05) of protein 53, Bcl-2 associated X protein (Bax), and apoptosis antigen 1 (Fas) were found in RG1 fetuses; and thymuses of RG2 fetuses had increased caspase-3, and expression of Fas and Bax (P < 0.05), relative to control fetuses. CONCLUSION These results indicate that reduced cell proliferation, oxidative stress, and increased cell apoptosis were the potential mechanisms for impaired development and microenvironment of IUGR fetal thymus, and for modifying the maturation of CD4(+) CD8(+) thymocytes underlying their reduced numbers .
Collapse
Affiliation(s)
- Yingchun Liu
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China.,Inner Mongolia Key Laboratory of Biomanufacturing, Hohhot, China
| | - Shan He
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuan Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Wei Xia
- College of Life Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Ming Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Chongzhi Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| | - Feng Gao
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
32
|
Abstract
Understanding a complex pathology such as inflammatory bowel disease, where host genetics (innate and adaptive immunity, barrier function) and environmental factors (microbes, diet, and stress) interact together to influence disease onset and severity, requires multipronged approaches to model these numerous variables. Researchers have typically relied on preclinical models of mouse and rat origin to push the boundary of knowledge further. However, incorporation of novel vertebrate models may contribute to new knowledge on specific aspects of intestinal homeostasis. An emerging literature has seen the use of zebrafish as a novel animal system to study key aspects of host-microbe interactions in the intestine. In this review, we briefly introduce components of host-microbiota interplay in the developing zebrafish intestine and summarize key lessons learned from this animal system; review important chemically induced and genetically engineered zebrafish models of intestinal immune disorders; and discuss perspectives and limitations of the zebrafish model system.
Collapse
Affiliation(s)
- Ye Yang
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Sarah Tomkovich
- Department of Medicine, University of Florida, Gainesville, Florida,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida,Department of Infectious Diseases and Pathology, University of Florida, Gainesville, Florida
| |
Collapse
|
33
|
Common Concepts of Immune Defense. Comp Med 2014. [DOI: 10.1007/978-3-7091-1559-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
34
|
Abstract
Age-related regression of the thymus is associated with a decline in naïve T cell output. This is thought to contribute to the reduction in T cell diversity seen in older individuals and linked with increased susceptibility to infection, autoimmune disease, and cancer. Thymic involution is one of the most dramatic and ubiquitous changes seen in the aging immune system, but the mechanisms which underlying this process are poorly understood. However, a picture is emerging, implicating the involvement of both extrinsic and intrinsic factors. In this review we assess the role of the thymic microenvironment as a potential target that regulates thymic involution, question whether thymocyte development in the aged thymus is functionally impaired, and explore the kinetics of thymic involution.
Collapse
Affiliation(s)
- Donald B Palmer
- Infection and Immunity Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, University of London , London , UK
| |
Collapse
|
35
|
Pantic I, Pantic S, Paunovic J, Perovic M. Nuclear entropy, angular second moment, variance and texture correlation of thymus cortical and medullar lymphocytes: grey level co-occurrence matrix analysis. AN ACAD BRAS CIENC 2013; 85:1063-72. [PMID: 23969846 DOI: 10.1590/s0001-37652013005000045] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 10/11/2012] [Indexed: 02/05/2023] Open
Abstract
Grey level co-occurrence matrix analysis (GLCM) is a well-known mathematical method for quantification of cell and tissue textural properties, such as homogeneity, complexity and level of disorder. Recently, it was demonstrated that this method is capable of evaluating fine structural changes in nuclear structure that otherwise are undetectable during standard microscopy analysis. In this article, we present the results indicating that entropy, angular second moment, variance, and texture correlation of lymphocyte nuclear structure determined by GLCM method are different in thymus cortex when compared to medulla. A total of 300 thymus lymphocyte nuclei from 10 one-month-old mice were analyzed: 150 nuclei from cortex and 150 nuclei from medullar regions of thymus. Nuclear GLCM analysis was carried out using National Institutes of Health ImageJ software. For each nucleus, entropy, angular second moment, variance and texture correlation were determined. Cortical lymphocytes had significantly higher chromatin angular second moment (p < 0.001) and texture correlation (p < 0.05) compared to medullar lymphocytes. Nuclear GLCM entropy and variance of cortical lymphocytes were on the other hand significantly lower than in medullar lymphocytes (p < 0.001). These results suggest that GLCM as a method might have a certain potential in detecting discrete changes in nuclear structure associated with lymphocyte migration and maturation in thymus.
Collapse
Affiliation(s)
- Igor Pantic
- Laboratory for Cellular Physiology, School of Medicine, Institute of Medical Physiology, University of Belgrade, 11129 Belgrade, Serbia.
| | | | | | | |
Collapse
|
36
|
The critical role of STIM1-dependent Ca2+ signalling during T-cell development and activation. Int J Biochem Cell Biol 2013; 45:2491-5. [PMID: 23906672 DOI: 10.1016/j.biocel.2013.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 07/10/2013] [Accepted: 07/19/2013] [Indexed: 11/22/2022]
Abstract
T lymphocytes are key cellular effectors of adaptive immunity able to recognize a virtually limitless number of antigenic peptides and mount an immune response. Ca(2+) signals are crucial to the development and activation of T cells and Stromal Interaction Molecule 1 (STIM1) has been identified as a critical modulator of intracellular Ca(2+) levels in T cells. Although the role of STIM1 in T cell activation has been extensively investigated, the role of STIM1 in T cell development has been somewhat controversial. Indeed, deficiencies in STIM1 expression and function lead to both developmental defects associated with the development of autoimmunity yet also interfere with T cell activation leading to severe combined immunodeficiency signifying a multifaceted role of STIM1 in T cell physiology and pathophysiology.
Collapse
|
37
|
Foxn1 maintains thymic epithelial cells to support T-cell development via mcm2 in zebrafish. Proc Natl Acad Sci U S A 2012; 109:21040-5. [PMID: 23213226 DOI: 10.1073/pnas.1217021110] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The thymus is mainly comprised of thymic epithelial cells (TECs), which form the unique thymic epithelial microenvironment essential for intrathymic T-cell development. Foxn1, a member of the forkhead transcription factor family, is required for establishing a functional thymic rudiment. However, the molecular mechanisms underlying the function of Foxn1 are still largely unclear. Here, we show that Foxn1 functions in thymus development through Mcm2 in the zebrafish. We demonstrate that, in foxn1 knockdown embryos, the thymic rudiment is reduced and T-cell development is impaired. Genome-wide expression profiling shows that a number of genes, including some known thymopoiesis genes, are dysregulated during the initiation of the thymus primordium and immigration of T-cell progenitors to the thymus. Functional and epistatic studies show that mcm2 and cdca7 are downstream of Foxn1, and mcm2 is a direct target gene of Foxn1 in TECs. Finally, we find that the thymus defects in foxn1 and mcm2 morphants might be attributed to reduced cell proliferation rather than apoptosis. Our results reveal that the foxn1-mcm2 axis plays a central role in the genetic regulatory network controlling thymus development in zebrafish.
Collapse
|