1
|
Wu T, Chen S, Zhu X, Ma J, Luo M, Wang Y, Tian Y, Sun Q, Guo X, Zhang J, Zhang X, Zhu Y, Wu L. Dynamic regulation of innate lymphoid cell development during ontogeny. Mucosal Immunol 2024; 17:1285-1300. [PMID: 39159846 DOI: 10.1016/j.mucimm.2024.08.009] [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: 06/13/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
The helper-like ILC contains various functional subsets, such as ILC1, ILC2, ILC3 and LTi cells, mediating the immune responses against viruses, parasites, and extracellular bacteria, respectively. Among them, LTi cells are also crucial for the formation of peripheral lymphoid tissues, such as lymph nodes. Our research, along with others', indicates a high proportion of LTi cells in the fetal ILC pool, which significantly decreases after birth. Conversely, the proportion of non-LTi ILCs increases postnatally, corresponding to the need for LTi cells to mediate lymphoid tissue formation during fetal stages and other ILC subsets to combat diverse pathogen infections postnatally. However, the regulatory mechanism for this transition remains unclear. In this study, we observed a preference for fetal ILC progenitors to differentiate into LTi cells, while postnatal bone marrow ILC progenitors preferentially differentiate into non-LTi ILCs. Particularly, this differentiation shift occurs within the first week after birth in mice. Further analysis revealed that adult ILC progenitors exhibit stronger activation of the Notch signaling pathway compared to fetal counterparts, accompanied by elevated Gata3 expression and decreased Rorc expression, leading to a transition from fetal LTi cell-dominant states to adult non-LTi ILC-dominant states. This study suggests that the body can regulate ILC development by modulating the activation level of the Notch signaling pathway, thereby acquiring different ILC subsets to accommodate the varying demands within the body at different developmental stages.
Collapse
Affiliation(s)
- Tao Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Science, Beijing, China
| | - Sijie Chen
- MOE Key Lab of Bioinformatics/Bioinformatics Division, BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xinyi Zhu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Science, Beijing, China
| | - Jie Ma
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Maocai Luo
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yuanhao Wang
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Yujie Tian
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Qingqing Sun
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Xiaohuan Guo
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Jianhong Zhang
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Xuegong Zhang
- MOE Key Lab of Bioinformatics/Bioinformatics Division, BNRIST and Department of Automation, Tsinghua University, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Yunping Zhu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Li Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Science, Beijing, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
2
|
Huyghe M, Desterke C, Imeri J, Belliard N, Chaker D, Oudrirhi N, Bezerra H, Turhan AG, Bennaceur-Griscelli A, Griscelli F. Comparative analysis of iPSC-derived NK cells from two differentiation strategies reveals distinct signatures and cytotoxic activities. Front Immunol 2024; 15:1463736. [PMID: 39445004 PMCID: PMC11496199 DOI: 10.3389/fimmu.2024.1463736] [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: 07/12/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
Purpose The ability to generate natural killer (NK) cells from induced pluripotent stem cells (iPSCs) has given rise to new possibilities for the large-scale production of homogeneous immunotherapeutic cellular products and opened new avenues towards the creation of "off-the-shelf" cancer immunotherapies. However, the differentiation of NK cells from iPSCs remains poorly understood, particularly regarding the ontogenic landscape of iPSC-derived NK (iNK) cells produced in vitro and the influence that the differentiation strategy employed may have on the iNK profile. Methods To investigate this question, we conducted a comparative analysis of two sets of iNK cells generated from the same iPSC line using two different protocols: (i) a short-term, clinically compatible feeder-free protocol corresponding to primitive hematopoiesis, and (ii) a lymphoid-based protocol representing the definitive hematopoietic step. Results and discussion Our work demonstrated that both protocols are capable of producing functional iNK cells. However, the two sets of resulting iNKs exhibited distinct phenotypes and transcriptomic profiles. The lymphoid-based differentiation approach generated iNKs with a more mature and activated profile, which demonstrated higher cytotoxicity against cancer cell lines compared to iNK cells produced under short-term feeder-free conditions suggesting that the differentiation strategy must be considered when designing iNK cell-based adoptive immunotherapies.
Collapse
Affiliation(s)
- Matthias Huyghe
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
| | - Christophe Desterke
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
| | - Jusuf Imeri
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
| | - Nathan Belliard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
| | - Diana Chaker
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
- Unités Mixtes de Service (UMS 045)- CITHERA (Center for iPSC Cell Therapy), National Infrastructure INGESTEM, Corbeil-Essonnes, Evry, France
| | - Noufissa Oudrirhi
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
- Service d’Hématologie Biologique Unité d’Onco-Hématologie moléculaire et Cytogénétique Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Universitaire Paris Sud Paul-Brousse, Villejuif, France
| | - Hudson Bezerra
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
| | - Ali G. Turhan
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
- Unités Mixtes de Service (UMS 045)- CITHERA (Center for iPSC Cell Therapy), National Infrastructure INGESTEM, Corbeil-Essonnes, Evry, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France
| | - Annelise Bennaceur-Griscelli
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
- Unités Mixtes de Service (UMS 045)- CITHERA (Center for iPSC Cell Therapy), National Infrastructure INGESTEM, Corbeil-Essonnes, Evry, France
- Service d’Hématologie Biologique Unité d’Onco-Hématologie moléculaire et Cytogénétique Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Universitaire Paris Sud Paul-Brousse, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Kremlin Bicêtre, France
| | - Frank Griscelli
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR-S-1310), Villejuif, France
- Unités Mixtes de Service (UMS 045)- CITHERA (Center for iPSC Cell Therapy), National Infrastructure INGESTEM, Corbeil-Essonnes, Evry, France
- Université Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
- Institut Gustave-Roussy, Département de Biologie et Pathologie Médicale, Villejuif, France
| |
Collapse
|
3
|
Shin B, Chang SJ, MacNabb BW, Rothenberg EV. Transcriptional network dynamics in early T cell development. J Exp Med 2024; 221:e20230893. [PMID: 39167073 PMCID: PMC11338287 DOI: 10.1084/jem.20230893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/07/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
The rate at which cells enter the T cell pathway depends not only on the immigration of hematopoietic precursors into the strong Notch signaling environment of the thymus but also on the kinetics with which each individual precursor cell reaches T-lineage commitment once it arrives. Notch triggers a complex, multistep gene regulatory network in the cells in which the steps are stereotyped but the transition speeds between steps are variable. Progenitor-associated transcription factors delay T-lineage differentiation even while Notch-induced transcription factors within the same cells push differentiation forward. Progress depends on regulator cross-repression, on breaching chromatin barriers, and on shifting, competitive collaborations between stage-specific and stably expressed transcription factors, as reviewed here.
Collapse
Affiliation(s)
- Boyoung Shin
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Samantha J Chang
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Brendan W MacNabb
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| |
Collapse
|
4
|
Das A, Martinez-Ruiz GU, Bouladoux N, Stacy A, Moraly J, Vega-Sendino M, Zhao Y, Lavaert M, Ding Y, Morales-Sanchez A, Harly C, Seedhom MO, Chari R, Awasthi P, Ikeuchi T, Wang Y, Zhu J, Moutsopoulos NM, Chen W, Yewdell JW, Shapiro VS, Ruiz S, Taylor N, Belkaid Y, Bhandoola A. Transcription factor Tox2 is required for metabolic adaptation and tissue residency of ILC3 in the gut. Immunity 2024; 57:1019-1036.e9. [PMID: 38677292 PMCID: PMC11096055 DOI: 10.1016/j.immuni.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/13/2024] [Accepted: 04/03/2024] [Indexed: 04/29/2024]
Abstract
Group 3 innate lymphoid cells (ILC3) are the major subset of gut-resident ILC with essential roles in infections and tissue repair, but how they adapt to the gut environment to maintain tissue residency is unclear. We report that Tox2 is critical for gut ILC3 maintenance and function. Gut ILC3 highly expressed Tox2, and depletion of Tox2 markedly decreased ILC3 in gut but not at central sites, resulting in defective control of Citrobacter rodentium infection. Single-cell transcriptional profiling revealed decreased expression of Hexokinase-2 in Tox2-deficient gut ILC3. Consistent with the requirement for hexokinases in glycolysis, Tox2-/- ILC3 displayed decreased ability to utilize glycolysis for protein translation. Ectopic expression of Hexokinase-2 rescued Tox2-/- gut ILC3 defects. Hypoxia and interleukin (IL)-17A each induced Tox2 expression in ILC3, suggesting a mechanism by which ILC3 adjusts to fluctuating environments by programming glycolytic metabolism. Our results reveal the requirement for Tox2 to support the metabolic adaptation of ILC3 within the gastrointestinal tract.
Collapse
Affiliation(s)
- Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Gustavo Ulises Martinez-Ruiz
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA; Faculty of Medicine, Research Division, National Autonomous University of Mexico, Mexico City, Mexico; Children's Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Nicolas Bouladoux
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA
| | - Apollo Stacy
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA; Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Josquin Moraly
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Maria Vega-Sendino
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Yongge Zhao
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Yi Ding
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Abigail Morales-Sanchez
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA; Children's Hospital of Mexico Federico Gomez, Mexico City, Mexico
| | - Christelle Harly
- Université de Nantes, CNRS, Inserm, CRCINA, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology," Nantes, France
| | - Mina O Seedhom
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Parirokh Awasthi
- Mouse Modeling Core, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Tomoko Ikeuchi
- Oral Immunity and Infection Section, NIDCR, NIH, Bethesda, MD, USA
| | - Yueqiang Wang
- Shenzhen Typhoon HealthCare, Shenzhen, Guangdong, China
| | - Jinfang Zhu
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | | | - WanJun Chen
- Mucosal Immunology Section, NIDCR, NIH, Bethesda, MD, USA
| | | | | | - Sergio Ruiz
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, NIAID, NIH, Bethesda, MD, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, NCI, NIH, Bethesda, MD, USA.
| |
Collapse
|
5
|
Li L, Shi J, Liu W, Luo Y, Gao S, Liu JX. Copper overload induces apoptosis and impaired proliferation of T cell in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2024; 267:106808. [PMID: 38159456 DOI: 10.1016/j.aquatox.2023.106808] [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: 07/20/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024]
Abstract
Copper is an essential biometal for cell development and function, however, unbalanced copper homeostasis in T cell development and the underlying mechanisms are largely unexplored. Here, we use a zebrafish model to investigate the effect of copper overload in T cell development. We show that copper stressed zebrafish larvae exhibit a significant reduction in T cells with increased cell apoptosis and impaired cell proliferation. T cell progenitors, hematopoietic stem and progenitor cells, also exhibit increased cell apoptosis. Copper overload induces production of ROS and the down-regulations of its resistance genes foxos, and ectopic expression of foxo3a, ROS scavenger GSH, could both effectively rescue the reduction of T cells in copper overload larvae. Moreover, foxm1-cytoskeleton axis, parallel to ROS-foxo axis, also mediates the copper overload induced T cell developmental defects. Meanwhile, ROS destroys expression of cytoskeleton rather than of foxm1 in the cells to induce cell apoptosis and the impaired proliferation. The functional integrity of copper transporters cox17 and atp7b are required for copper stress in inducing T cell apoptosis and proliferation impairment. Our findings demonstrate that the down-stream ROS-foxo/cytoskeleton and foxm1-cytoskeleton signaling pathways contribute jointly to copper overload induced T cell apoptosis and proliferation defects, which are depend on the integral function of Cox17 and Atp7b, and provide new insight into the copper homeostasis in T lymphocyte development.
Collapse
Affiliation(s)
- LingYa Li
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - JiaHao Shi
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - WenYe Liu
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yi Luo
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Sheng Gao
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jing-Xia Liu
- Key Laboratory of Freshwater Animal Breeding, College of Fisheries, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
6
|
Maruyama S. The Functional Assessment of T Cells. Methods Mol Biol 2024; 2766:207-232. [PMID: 38270882 DOI: 10.1007/978-1-0716-3682-4_23] [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] [Indexed: 01/26/2024]
Abstract
It is important to know what kind of T-cell populations are involved in various disease states, and to know the state of T-cell functions involving in the disease. When a T cell's antigen receptors (TCR) recognize a specific antigen, the cell transmits a signal by a transduction mechanism within the T cell's cytoplasm. This signal initiates gene transcription essential for differentiation and activation of T cells. In this chapter, we will describe the methods of analyzing the transcribed mRNA and detecting the translated product in order to know the activation state of T cells.
Collapse
Affiliation(s)
- Saho Maruyama
- Department of Basic Medical Research and Education, Ehime University Graduate School of Medicine, Toon, Ehime, Japan.
| |
Collapse
|
7
|
Nishimura A, Yokoyama K, Naruto T, Yamagishi C, Imamura T, Nakazono H, Kimura S, Ito M, Sagisaka M, Tanaka Y, Piao J, Namikawa Y, Yanagimachi M, Isoda T, Kanai A, Matsui H, Isobe T, Sato-Otsubo A, Higuchi N, Takada A, Okuno H, Saito S, Karakawa S, Kobayashi S, Hasegawa D, Fujisaki H, Hasegawa D, Koike K, Koike T, Rai S, Umeda K, Sano H, Sekinaka Y, Ogawa A, Kinoshita A, Shiba N, Miki M, Kimura F, Nakayama H, Nakazawa Y, Taga T, Taki T, Adachi S, Manabe A, Koh K, Ishida Y, Takita J, Ishikawa F, Goto H, Morio T, Mizutani S, Tojo A, Takagi M. Myeloid/natural killer (NK) cell precursor acute leukemia as a distinct leukemia type. SCIENCE ADVANCES 2023; 9:eadj4407. [PMID: 38091391 PMCID: PMC10848711 DOI: 10.1126/sciadv.adj4407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023]
Abstract
Myeloid/natural killer (NK) cell precursor acute leukemia (MNKPL) has been described on the basis of its unique immunophenotype and clinical phenotype. However, there is no consensus on the characteristics for identifying this disease type because of its rarity and lack of defined distinctive molecular characteristics. In this study, multiomics analysis revealed that MNKPL is distinct from acute myeloid leukemia, T cell acute lymphoblastic leukemia, and mixed-phenotype acute leukemia (MPAL), and NOTCH1 and RUNX3 activation and BCL11B down-regulation are hallmarks of MNKPL. Although NK cells have been classically considered to be lymphoid lineage-derived, the results of our single-cell analysis using MNKPL cells suggest that NK cells and myeloid cells share common progenitor cells. Treatment outcomes for MNKPL are unsatisfactory, even when hematopoietic cell transplantation is performed. Multiomics analysis and in vitro drug sensitivity assays revealed increased sensitivity to l-asparaginase and reduced levels of asparagine synthetase (ASNS), supporting the clinically observed effectiveness of l-asparaginase.
Collapse
Affiliation(s)
- Akira Nishimura
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuaki Yokoyama
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takuya Naruto
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chika Yamagishi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshihiko Imamura
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
| | - Hiroto Nakazono
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shunsuke Kimura
- Department of Pediatrics, Hiroshima University Graduate School of Biomedical Sciences, Hiroshima, Japan
| | - Mieko Ito
- Division of Hematology/Oncology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Maiko Sagisaka
- Division of Hematology/Oncology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Yukie Tanaka
- Research Core, Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Jinhua Piao
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yui Namikawa
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masakatsu Yanagimachi
- Division of Hematology/Oncology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Takeshi Isoda
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Akinori Kanai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Hirotaka Matsui
- Department of Molecular Laboratory Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoya Isobe
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aiko Sato-Otsubo
- Department of Pediatrics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoko Higuchi
- Department of Pediatrics, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akiko Takada
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruna Okuno
- Department of Pediatrics, Gunma University Hospital, Maebashi, Japan
| | - Shoji Saito
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shuhei Karakawa
- Department of Pediatrics, Hiroshima University Hospital, Hiroshima, Japan
| | - Shogo Kobayashi
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Daisuke Hasegawa
- Department of Pediatrics, St. Luke’s International Hospital, Tokyo, Japan
| | - Hiroyuki Fujisaki
- Department of Pediatric Hematology and Oncology, Osaka City General Hospital, Osaka, Japan
| | - Daiichiro Hasegawa
- Department of Hematology and Oncology, Hyogo Prefectural Kobe Children’s Hospital, Kobe, Japan
| | - Kazutoshi Koike
- Division of Pediatric Hematology and Oncology, Ibaraki Children's Hospital, Mito, Japan
| | - Takashi Koike
- Department of Pediatrics, Tokai University School of Medicine, Isehara, Japan
| | - Shinya Rai
- Department of Hematology and Rheumatology, Faculty of Medicine, Kindai University, Osakasayama, Japan
| | - Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hideki Sano
- Department of Pediatric Oncology, Fukushima Medical University Hospital, Fukushima, Japan
| | - Yujin Sekinaka
- Department of Pediatrics, National Defense Medical College, Tokorozawa, Japan
| | - Atsushi Ogawa
- Department of Pediatrics, Niigata Cancer Center Hospital, Niigata, Japan
| | - Akitoshi Kinoshita
- Department of Pediatrics, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Norio Shiba
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mizuka Miki
- Department of Pediatrics, Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima, Japan
| | - Fumihiko Kimura
- Division of Hematology, Department of Internal Medicine, National Defense Medical College, Tokorozawa, Japan
| | - Hideki Nakayama
- Department of Pediatrics, Kyushu Cancer Center, Fukuoka, Japan
| | - Yozo Nakazawa
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takashi Taga
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Department of Pediatrics, Shiga University of Medical Science, Ohtsu, Japan
| | - Tomohiko Taki
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Department of Medical Technology, Faculty of Health Sciences, Kyorin University, Tokyo, Japan
| | - Souichi Adachi
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Department of Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Manabe
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Department of Pediatrics, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Katsuyoshi Koh
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Department of Hematology/Oncology, Saitama Children’s Medical Center, Saitama, Japan
| | - Yasushi Ishida
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
- Pediatric Medical Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Junko Takita
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Ishikawa
- Laboratory for Human Disease Models, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Comprehensive Pathology, Tokyo Medical and Dental University University (TMDU), Tokyo, Japan
| | - Hiroaki Goto
- Division of Hematology/Oncology, Kanagawa Children’s Medical Center, Yokohama, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shuki Mizutani
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Arinobu Tojo
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Data Science and Faculty Affairs, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masatoshi Takagi
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
- Leukemia/Lymphoma Committee of Japanese Society of Pediatric Hematology and Oncology, Tokyo, Japan
| |
Collapse
|
8
|
Cheng Y, Ren Y, Wang W, Zhang W. Similar proteome expression profiles of the aggregated lymphoid nodules area and Peyer's patches in Bactrian camel. BMC Genomics 2023; 24:608. [PMID: 37821839 PMCID: PMC10568864 DOI: 10.1186/s12864-023-09715-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The presence of Aggregated Lymphoid Nodules Area (ALNA) is a notable anatomical characteristic observed in the abomasum of Bactrian camels. This area is comprised of two separate regions, namely the Reticular Mucosal Folds Region (RMFR) and the Longitudinal Mucosal Folds Region (LMFR). The histological properties of ALNA exhibit significant similarities to those of Peyer's patches (PPs) found in the gastrointestinal system. The functional characteristics of ALNA were examined in relation to mucosal immunity in the gastrointestinal system. RESULTS We used iTRAQ-based proteomic analysis on twelve Bactrian camels to measure the amount of proteins expressed in ALNA. In the experiment, we sampled the RMFR and LMFR separately from the ALNA and compared their proteomic quantification results with samples from the PPs. A total of 1253 proteins were identified, among which 39 differentially expressed proteins (DEPs) were found between RMFR and PPs, 33 DEPs were found between LMFR and PPs, and 22 DEPs were found between LMFR and RMFR. The proteins FLNA, MYH11, and HSPB1 were chosen for validation using the enzyme-linked immunosorbent assay (ELISA), and the observed expression profiles were found to be in agreement with the results obtained from the iTRAQ study. The InnateDB database was utilized to get data pertaining to immune-associated proteins in ALNA. It was observed that a significant proportion, specifically 76.6%, of these proteins were found to be associated with the same orthogroups as human immune-related genes. These proteins are acknowledged to be associated with a diverse range of functions, encompassing the uptake, processing and presentation of antigens, activation of lymphocytes, the signaling pathways of T-cell and B-cell receptors, and the control of actin polymerization. CONCLUSIONS The experimental results suggest that there are parallels in the immune-related proteins found in ALNA and PPs. Although there are variations in the structures of LMFR and RMFR, the proteins produced in both structures exhibit a high degree of similarity and perform comparable functions in the context of mucosal immune responses.
Collapse
Affiliation(s)
- Yujiao Cheng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yan Ren
- The Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, 5371, Australia
| | - Wenhui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| | - Wangdong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
| |
Collapse
|
9
|
MacNabb BW, Rothenberg EV. Speed and navigation control of thymocyte development by the fetal T-cell gene regulatory network. Immunol Rev 2023; 315:171-196. [PMID: 36722494 PMCID: PMC10771342 DOI: 10.1111/imr.13190] [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: 02/02/2023]
Abstract
T-cell differentiation is a tightly regulated developmental program governed by interactions between transcription factors (TFs) and chromatin landscapes and affected by signals received from the thymic stroma. This process is marked by a series of checkpoints: T-lineage commitment, T-cell receptor (TCR)β selection, and positive and negative selection. Dynamically changing combinations of TFs drive differentiation along the T-lineage trajectory, through mechanisms that have been most extensively dissected in adult mouse T-lineage cells. However, fetal T-cell development differs from adult in ways that suggest that these TF mechanisms are not fully deterministic. The first wave of fetal T-cell differentiation occurs during a unique developmental window during thymic morphogenesis, shows more rapid kinetics of differentiation with fewer rounds of cell division, and gives rise to unique populations of innate lymphoid cells (ILCs) and invariant γδT cells that are not generated in the adult thymus. As the characteristic kinetics and progeny biases are cell-intrinsic properties of thymic progenitors, the differences could be based on distinct TF network circuitry within the progenitors themselves. Here, we review recent single-cell transcriptome data that illuminate the TF networks involved in T-cell differentiation in the fetal and adult mouse thymus.
Collapse
Affiliation(s)
- Brendan W MacNabb
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
10
|
Rich RR, Cron RQ. The Human Immune Response. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00001-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
11
|
Group 2 innate lymphoid cells in human asthma. Allergol Int 2022; 72:194-200. [PMID: 36585333 DOI: 10.1016/j.alit.2022.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/30/2022] Open
Abstract
Asthma is characterized by increased airway hyperresponsiveness, reversible airflow limitation, and remodeling due to allergic airway inflammation. Asthma has been proposed to be classified into various phenotypes by cluster analyses integrating clinical information and laboratory data. Recently, asthma has been classified into two major endotypes, Type 2-high and Type 2-low asthma, and various subtypes based on the underlying molecular mechanisms. In Type 2-high asthma, Th2 cells, together with group 2 innate lymphoid cells (ILC2s), produce type 2 cytokines such as IL-4, IL-5, IL-9, and IL-13, which play crucial roles in causing airway inflammation. The roles of ILC2s in asthma pathogenesis have been analyzed primarily in murine models, demonstrating their importance not only in IL-33- or papain-induced innate asthma models but also in house dust mite (HDM)- or ovalbumin (OVA)-induced acquired asthma models evoked in an antigen-specific manner. Recently, evidence regarding the roles of ILC2s in human asthma is also accumulating. This minireview summarizes the roles of ILC2s in asthma, emphasizing human studies.
Collapse
|
12
|
Beckstette M, Lu CW, Herppich S, Diem EC, Ntalli A, Ochel A, Kruse F, Pietzsch B, Neumann K, Huehn J, Floess S, Lochner M. Profiling of epigenetic marker regions in murine ILCs under homeostatic and inflammatory conditions. J Exp Med 2022; 219:213389. [PMID: 35938981 PMCID: PMC9386974 DOI: 10.1084/jem.20210663] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/26/2022] [Accepted: 07/14/2022] [Indexed: 12/03/2022] Open
Abstract
Epigenetic modifications such as DNA methylation play an essential role in imprinting specific transcriptional patterns in cells. We performed genome-wide DNA methylation profiling of murine lymph node–derived ILCs, which led to the identification of differentially methylated regions (DMRs) and the definition of epigenetic marker regions in ILCs. Marker regions were located in genes with a described function for ILCs, such as Tbx21, Gata3, or Il23r, but also in genes that have not been related to ILC biology. Methylation levels of the marker regions and expression of the associated genes were strongly correlated, indicating their functional relevance. Comparison with T helper cell methylomes revealed clear lineage differences, despite partial similarities in the methylation of specific ILC marker regions. IL-33–mediated challenge affected methylation of ILC2 epigenetic marker regions in the liver, while remaining relatively stable in the lung. In our study, we identified a set of epigenetic markers that can serve as a tool to study phenotypic and functional properties of ILCs.
Collapse
Affiliation(s)
- Michael Beckstette
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Bielefeld Institute for Bioinformatics Infrastructure, Department of Technology, Bielefeld University, Bielefeld, Germany
| | - Chia-Wen Lu
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.,Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Susanne Herppich
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Elia C Diem
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Anna Ntalli
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Aaron Ochel
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Kruse
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hannover, Germany
| | - Beate Pietzsch
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jochen Huehn
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Stefan Floess
- Department Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Matthias Lochner
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.,Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hannover, Germany
| |
Collapse
|
13
|
Meermeier EW, Zheng CL, Tran JG, Soma S, Worley AH, Weiss DI, Modlin RL, Swarbrick G, Karamooz E, Khuzwayo S, Wong EB, Gold MC, Lewinsohn DM. Human lung-resident mucosal-associated invariant T cells are abundant, express antimicrobial proteins, and are cytokine responsive. Commun Biol 2022; 5:942. [PMID: 36085311 PMCID: PMC9463188 DOI: 10.1038/s42003-022-03823-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
Mucosal-associated Invariant T (MAIT) cells are an innate-like T cell subset that recognize a broad array of microbial pathogens, including respiratory pathogens. Here we investigate the transcriptional profile of MAIT cells localized to the human lung, and postulate that MAIT cells may play a role in maintaining homeostasis at this mucosal barrier. Using the MR1/5-OP-RU tetramer, we identified MAIT cells and non-MAIT CD8+ T cells in lung tissue not suitable for transplant from human donors. We used RNA-sequencing of MAIT cells compared to non-MAIT CD8+ T cells to define the transcriptome of MAIT cells in the human lung. We show that, as a population, lung MAIT cells are polycytotoxic, secrete the directly antimicrobial molecule IL-26, express genes associated with persistence, and selectively express cytokine and chemokine- related molecules distinct from other lung-resident CD8+ T cells, such as interferon-γ- and IL-12- receptors. These data highlight MAIT cells' predisposition to rapid pro-inflammatory cytokine responsiveness and antimicrobial mechanisms in human lung tissue, concordant with findings of blood-derived counterparts, and support a function for MAIT cells as early sensors in the defense of respiratory barrier function.
Collapse
Affiliation(s)
- Erin W Meermeier
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Christina L Zheng
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jessica G Tran
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Shogo Soma
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aneta H Worley
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - David I Weiss
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Gwendolyn Swarbrick
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Elham Karamooz
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Sharon Khuzwayo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Emily B Wong
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Infection and Immunity, University College London, London, UK
| | - Marielle C Gold
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - David M Lewinsohn
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
14
|
Pankow A, Sun XH. The divergence between T cell and innate lymphoid cell fates controlled by E and Id proteins. Front Immunol 2022; 13:960444. [PMID: 36032069 PMCID: PMC9399370 DOI: 10.3389/fimmu.2022.960444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/13/2022] [Indexed: 11/18/2022] Open
Abstract
T cells develop in the thymus from lymphoid primed multipotent progenitors or common lymphoid progenitors into αβ and γδ subsets. The basic helix-loop-helix transcription factors, E proteins, play pivotal roles at multiple stages from T cell commitment to maturation. Inhibitors of E proteins, Id2 and Id3, also regulate T cell development while promoting ILC differentiation. Recent findings suggest that the thymus can also produce innate lymphoid cells (ILCs). In this review, we present current findings that suggest the balance between E and Id proteins is likely to be critical for controlling the bifurcation of T cell and ILC fates at early stages of T cell development.
Collapse
Affiliation(s)
- Aneta Pankow
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Xiao-Hong Sun
- Program in Arthritis and Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Xiao-Hong Sun,
| |
Collapse
|
15
|
Ren G, Lai B, Harly C, Baek S, Ding Y, Zheng M, Cao Y, Cui K, Yang Y, Zhu J, Hager GL, Bhandoola A, Zhao K. Transcription factors TCF-1 and GATA3 are key factors for the epigenetic priming of early innate lymphoid progenitors toward distinct cell fates. Immunity 2022; 55:1402-1413.e4. [PMID: 35882235 PMCID: PMC9393082 DOI: 10.1016/j.immuni.2022.06.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 11/17/2022]
Abstract
The differentiation of innate lymphoid cells (ILCs) from hematopoietic stem cells needs to go through several multipotent progenitor stages. However, it remains unclear whether the fates of multipotent progenitors are predefined by epigenetic states. Here, we report the identification of distinct accessible chromatin regions in all lymphoid progenitors (ALPs), EILPs, and ILC precursors (ILCPs). Single-cell MNase-seq analyses revealed that EILPs contained distinct subpopulations epigenetically primed toward either dendritic cell lineages or ILC lineages. We found that TCF-1 and GATA3 co-bound to the lineage-defining sites for ILCs (LDS-Is), whereas PU.1 binding was enriched in the LDSs for alternative dendritic cells (LDS-As). TCF-1 and GATA3 were indispensable for the epigenetic priming of LDSs at the EILP stage. Our results suggest that the multipotency of progenitor cells is defined by the existence of a heterogeneous population of cells epigenetically primed for distinct downstream lineages, which are regulated by key transcription factors.
Collapse
Affiliation(s)
- Gang Ren
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; Northwest Agriculture and Forest University, College of Animal Science and Technology, Yangling, Shaanxi 712100, China
| | - Binbin Lai
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA; Biomedical Engineering Department, Peking University, Beijing 100191, China; Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
| | - Christelle Harly
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Ding
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mingzhu Zheng
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA; Department of Microbiology and Immunology, School of Medicine, Jiangsu Provincial Key Laboratory of Critical Care Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yaqiang Cao
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Kairong Cui
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Yu Yang
- Biomedical Engineering Department, Peking University, Beijing 100191, China; Department of Dermatology and Venereology, Peking University First Hospital, Beijing 100034, China
| | - Jinfang Zhu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keji Zhao
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Joyce S. Life In-Between: Bridging Innate and Adaptive Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:193-195. [PMID: 35821100 DOI: 10.4049/jimmunol.2290012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
|
17
|
Huseby ES, Teixeiro E. The perception and response of T cells to a changing environment are based on the law of initial value. Sci Signal 2022; 15:eabj9842. [PMID: 35639856 PMCID: PMC9290192 DOI: 10.1126/scisignal.abj9842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
αβ T cells are critical components of the adaptive immune system and are capable of inducing sterilizing immunity after pathogen infection and eliminating transformed tumor cells. The development and function of T cells are controlled through the T cell antigen receptor, which recognizes peptides displayed on major histocompatibility complex (MHC) molecules. Here, we review how T cells generate the ability to recognize self-peptide-bound MHC molecules and use signals derived from these interactions to instruct cellular development, activation thresholds, and functional specialization in the steady state and during immune responses. We argue that the basic tenants of T cell development and function follow Weber-Fetcher's law of just noticeable differences and Wilder's law of initial value. Together, these laws argue that the ability of a system to respond and the quality of that response are scalable to the basal state of that system. Manifestation of these laws in T cells generates clone-specific activation thresholds that are based on perceivable differences between homeostasis and pathogen encounter (self versus nonself discrimination), as well as poised states for subsequent differentiation into specific effector cell lineages.
Collapse
Affiliation(s)
- Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
18
|
Zhou W, Gao F, Romero-Wolf M, Jo S, Rothenberg EV. Single-cell deletion analyses show control of pro-T cell developmental speed and pathways by Tcf7, Spi1, Gata3, Bcl11a, Erg, and Bcl11b. Sci Immunol 2022; 7:eabm1920. [PMID: 35594339 PMCID: PMC9273332 DOI: 10.1126/sciimmunol.abm1920] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
As early T cell precursors transition from multipotentiality to T lineage commitment, they change expression of multiple transcription factors. It is unclear whether individual transcription factors directly control choices between T cell identity and some alternative fate or whether these factors mostly affect proliferation or survival during the normal commitment process. Here, we unraveled the impacts of deleting individual transcription factors at two stages in early T cell development, using synchronized in vitro differentiation systems, single-cell RNA-seq with batch indexing, and controlled gene-disruption strategies. First, using a customized method for single-cell CRISPR disruption, we defined how the early-acting transcription factors Bcl11a, Erg, Spi1 (PU.1), Gata3, and Tcf7 (TCF1) function before commitment. The results revealed a kinetic tug of war within individual cells between T cell factors Tcf7 and Gata3 and progenitor factors Spi1 and Bcl11a, with an unexpected guidance role for Erg. Second, we tested how activation of transcription factor Bcl11b during commitment altered ongoing cellular programs. In knockout cells where Bcl11b expression was prevented, the cells did not undergo developmental arrest, instead following an alternative path as T lineage commitment was blocked. A stepwise, time-dependent regulatory cascade began with immediate-early transcription factor activation and E protein inhibition, finally leading Bcl11b knockout cells toward exit from the T cell pathway. Last, gene regulatory networks of transcription factor cross-regulation were extracted from the single-cell transcriptome results, characterizing the specification network operating before T lineage commitment and revealing its links to both the Bcl11b knockout alternative network and the network consolidating T cell identity during commitment.
Collapse
Affiliation(s)
- Wen Zhou
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Program in Biochemistry and Molecular Biophysics, California Institute of Technology
- Current address: BillionToOne, Menlo Park, CA
| | - Fan Gao
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Caltech Bioinformatics Resource Center, Beckman Institute of Caltech
| | - Maile Romero-Wolf
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Current address: Center for Stem Cell Biology and Regenerative Medicine, University of Southern California
| | - Suin Jo
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Current address: Washington University of St. Louis
| | - Ellen V. Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
| |
Collapse
|
19
|
Mu P, Huo J, Li X, Li W, Li X, Ao J, Chen X. IL-2 Signaling Couples the MAPK and mTORC1 Axes to Promote T Cell Proliferation and Differentiation in Teleosts. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1616-1631. [PMID: 35321881 DOI: 10.4049/jimmunol.2100764] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/27/2022] [Indexed: 06/14/2023]
Abstract
IL-2 is a pleiotropic cytokine that is critical for T cell immunity. Although the IL-2-mediated regulation of T cell immunity in mammals is relatively well understood, it remains largely unknown whether and how IL-2 regulates T cell immunity in lower vertebrates. To address this knowledge gap, we investigated the role played by IL-2 in the regulation of T cell response, as well as the associated underlying mechanisms in a teleost fish, large yellow croaker (Larimichthys crocea). We found that large yellow croaker (L. crocea) IL-2 (LcIL-2) significantly promoted T cell proliferation both in vivo and in vitro; significantly induced the differentiation of Th1, Th2, regulatory T, and cytotoxic T cells while inhibiting Th17 differentiation; and participated in the elimination of invading pathogenic bacteria. Mechanistically, the binding of LcIL-2 to its heterotrimer receptor complex (LcIL-15Rα/LcIL-2Rβ/Lcγc) triggered the conserved JAK-STAT5 pathway, which in turn regulated the expression of genes involved in T cell expansion, differentiation, and biological function. The MAPK and mammalian target of rapamycin complex 1 (mTORC1) axes, which are involved in TCR-mediated signaling, were also required for LcIL-2-mediated T cell response. Collectively, our results demonstrated that fish IL-2 plays a comprehensive regulatory role in T cell response and highlighted the complex and delicate network regulating T cell-driven immune response. We propose that T cell immunity is regulated by the interplay between TCR signaling and cytokine signaling, and that this basic strategy evolved before the emergence of the tetrapod lineage. Our findings provide valuable insights into the regulatory mechanisms underlying T cell response in teleosts.
Collapse
Affiliation(s)
- Pengfei Mu
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China; and
| | - Jieying Huo
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China; and
| | - Xiaofeng Li
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Wanru Li
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaomeng Li
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jingqun Ao
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China; and
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, China;
- Key Laboratory of Marine Biogenetic Resources, Third Institute of Oceanography, Ministry of Natural Resources, Xiamen, China; and
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
20
|
Notch signaling pathway: architecture, disease, and therapeutics. Signal Transduct Target Ther 2022; 7:95. [PMID: 35332121 PMCID: PMC8948217 DOI: 10.1038/s41392-022-00934-y] [Citation(s) in RCA: 398] [Impact Index Per Article: 132.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
The NOTCH gene was identified approximately 110 years ago. Classical studies have revealed that NOTCH signaling is an evolutionarily conserved pathway. NOTCH receptors undergo three cleavages and translocate into the nucleus to regulate the transcription of target genes. NOTCH signaling deeply participates in the development and homeostasis of multiple tissues and organs, the aberration of which results in cancerous and noncancerous diseases. However, recent studies indicate that the outcomes of NOTCH signaling are changeable and highly dependent on context. In terms of cancers, NOTCH signaling can both promote and inhibit tumor development in various types of cancer. The overall performance of NOTCH-targeted therapies in clinical trials has failed to meet expectations. Additionally, NOTCH mutation has been proposed as a predictive biomarker for immune checkpoint blockade therapy in many cancers. Collectively, the NOTCH pathway needs to be integrally assessed with new perspectives to inspire discoveries and applications. In this review, we focus on both classical and the latest findings related to NOTCH signaling to illustrate the history, architecture, regulatory mechanisms, contributions to physiological development, related diseases, and therapeutic applications of the NOTCH pathway. The contributions of NOTCH signaling to the tumor immune microenvironment and cancer immunotherapy are also highlighted. We hope this review will help not only beginners but also experts to systematically and thoroughly understand the NOTCH signaling pathway.
Collapse
|
21
|
Abstract
TCF1 and its homologue LEF1 are historically known as effector transcription factors downstream of the WNT signalling pathway and are essential for early T cell development. Recent advances bring TCF1 into the spotlight for its versatile, context-dependent functions in regulating mature T cell responses. In the cytotoxic T cell lineages, TCF1 is required for the self-renewal of stem-like CD8+ T cells generated in response to viral or tumour antigens, and for preserving heightened responses to checkpoint blockade immunotherapy. In the helper T cell lineages, TCF1 is indispensable for the differentiation of T follicular helper and T follicular regulatory cells, and crucially regulates immunosuppressive functions of regulatory T cells. Mechanistic investigations have also identified TCF1 as the first transcription factor that directly modifies histone acetylation, with the capacity to bridge transcriptional and epigenetic regulation. TCF1 also has the potential to become an important clinical biomarker for assessing the prognosis of tumour immunotherapy and the success of viral control in treating HIV and hepatitis C virus infection. Here, we summarize the key findings on TCF1 across the fields of T cell immunity and reflect on the possibility of exploring TCF1 and its downstream transcriptional programmes as therapeutic targets for improving antiviral and antitumour immunity.
Collapse
|
22
|
Correlation between circulating innate lymphoid cell precursors and thymic function. iScience 2022; 25:103732. [PMID: 35118353 PMCID: PMC8792071 DOI: 10.1016/j.isci.2022.103732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/30/2021] [Accepted: 01/02/2022] [Indexed: 12/13/2022] Open
Abstract
The thymus has a high capacity to support the differentiation of ILCs, especially when E protein transcription factors are ablated. Whether it contributes to the homeostasis of ILC pools in tissues is not clear. Single-cell RNA sequencing analysis shows a substantial amount of ILC precursors in wild type but not athymic nude blood. The precursors express CD3 intracellularly (ic) but not on the surface. The abundance of Lin−CD127+CD62L+icCD3ε+ precursors varies with age, peaking at 2–3 months. These cells can differentiate into various ILC subsets on OP9-DL1 stroma in vitro. In the lung, small intestine, and epidermis, icCD3ε+ cells differentiate into diverse ILC subsets in different tissue environments in steady state. Helminth infection promotes their differentiation toward functional ILC2s. Thus, the thymus appears to play a role in replenishing ILC pools in different peripheral tissues. Because thymic activity is age-dependent, this finding may help explain age-related differences in immune responses. Single-cell RNA sequencing detects thymus-dependent (td) ILC precursors in the blood Intracellular (ic) but not surface CD3ε marks td-ILCs in the blood and tissues Blood td-ILCs differentiate into distinct ILC subsets in vitro Helminth infection promotes the maturation of icCD3ε+ ILC2s
Collapse
|
23
|
Temporal Gene Expression Profiles Reflect the Dynamics of Lymphoid Differentiation. Int J Mol Sci 2022; 23:ijms23031115. [PMID: 35163045 PMCID: PMC8834919 DOI: 10.3390/ijms23031115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/10/2022] [Accepted: 01/16/2022] [Indexed: 02/01/2023] Open
Abstract
Understanding the emergence of lymphoid committed cells from multipotent progenitors (MPP) is a great challenge in hematopoiesis. To gain deeper insight into the dynamic expression changes associated with these transitions, we report the quantitative transcriptome of two MPP subsets and the common lymphoid progenitor (CLP). While the transcriptome is rather stable between MPP2 and MPP3, expression changes increase with differentiation. Among those, we found that pioneer lymphoid genes such as Rag1, Mpeg1, and Dntt are expressed continuously from MPP2. Others, such as CD93, are CLP specific, suggesting their potential use as new markers to improve purification of lymphoid populations. Notably, a six-transcription factor network orchestrates the lymphoid differentiation program. Additionally, we pinpointed 24 long intergenic-non-coding RNA (lincRNA) differentially expressed through commitment and further identified seven novel forms. Collectively, our approach provides a comprehensive landscape of coding and non-coding transcriptomes expressed during lymphoid commitment.
Collapse
|
24
|
Das A, Harly C, Ding Y, Bhandoola A. ILC Differentiation from Progenitors in the Bone Marrow. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:7-24. [DOI: 10.1007/978-981-16-8387-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
25
|
ILC Differentiation in the Thymus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:25-39. [DOI: 10.1007/978-981-16-8387-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Ni G, Liu X, Li H, Fogarty CE, Chen S, Zhang P, Liu Y, Wu X, Wei MQ, Chen G, Zhang P, Wang T. Topical Application of Temperature-Sensitive Gel Containing Caerin 1.1 and 1.9 Peptides on TC-1 Tumour-Bearing Mice Induced High-Level Immune Response in the Tumour Microenvironment. Front Oncol 2021; 11:754770. [PMID: 34858827 PMCID: PMC8632150 DOI: 10.3389/fonc.2021.754770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 10/11/2021] [Indexed: 01/22/2023] Open
Abstract
The development of topical cream drugs that increase the immune activation of tumour-infiltrating lymphocytes against tumour and chronic viral infection-associated lesions is of great immunotherapeutic significance. This study demonstrates that the topical application of a temperature-sensitive gel containing caerin 1.1 and 1.9 peptides reduces nearly 50% of the tumour weight of HPV16 E6/E7-transformed TC-1 tumour-bearing mice via improving the tumour microenvironment. Confocal microscopy confirms the time-dependent penetration of caerin 1.9 through the epidermal layer of the ear skin structure of mice. Single-cell transcriptomic analysis shows that the caerin 1.1/1.9 gel expands the populations with high immune activation level and largely stimulates the pro-inflammatory activity of NK and dendritic cells. Closely associated with INFα response, Cebpb seems to play a key role in altering the function of all Arg1hi macrophages in the caerin group. In addition, the caerin gel treatment recruits almost two-fold more activated CD8+ T cells to the TME, relative to the untreated tumour, which shows a synergistic effect derived from the regulation of S1pr1, Ccr7, Ms4a4b and Gimap family expression. The TMT10plex-labelling proteomic quantification further demonstrates the activation of interferon-alpha/beta secretion and response to cytokine stimulus by the caerin gel, while the protein contents of several key regulators were elevated by more than 30%, such as Cd5l, Gzma, Ifit1, Irf9 and Stat1. Computational integration of the proteome with the single-cell transcriptome consistently suggested greater activation of NK and T cells with the topical application of caerin peptide gel.
Collapse
Affiliation(s)
- Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China.,Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia.,The First Affiliated Hospital/School of Clinical Medicine of Guangdong Pharmaceutical University , Guangzhou, China
| | - Xiaosong Liu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China.,Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Hejie Li
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Conor E Fogarty
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| | - Shu Chen
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Pingping Zhang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ying Liu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Xiaolian Wu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ming Q Wei
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Guoqiang Chen
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, China
| | - Ping Zhang
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, Australia
| | - Tianfang Wang
- Genecology Research Centre, University of the Sunshine Coast, Maroochydore DC, QLD, Australia.,School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, QLD, Australia
| |
Collapse
|
27
|
Wang C, Ruan L, Shi H, Lin W, Liu L, Li S. Phosphorylation of Shrimp Tcf by a Viral Protein Kinase WSV083 Suppresses Its Antiviral Effect. Front Immunol 2021; 12:698697. [PMID: 34408747 PMCID: PMC8365339 DOI: 10.3389/fimmu.2021.698697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022] Open
Abstract
Nuclear DNA-binding TCF proteins, which act as the main downstream effectors of Wnt signaling, are essential for the regulation of cell fate and innate immunity. However, their role during viral infection in shrimp remains unknown. Herein, we demonstrated that Litopenaeus vannamei TCF (LvTcf) acts independently of Lvβ-catenin to promote interferon-like protein LvVago1 production, thus mounting the response to WSSV infection. Further, we observed that WSV083, a WSSV serine/threonine protein kinase, bound to LvTcf and phosphorylated it. Phosphorylated LvTcf was then recognized and degraded via the ubiquitin-proteasome pathway. Moreover, mass spectrometry analyses indicated that the T39 and T104 residues of LvTcf were target sites phosphorylated by WSV083. Point mutation analyses suggested that additional sites of LvTcf may undergo phosphorylation via WSV083. Taken together, the current work provides valuable insights into host immunity and viral pathogenesis. LvTcf is not only a modulator of shrimp innate immunity but is also an important target for WSSV immune evasion. Thus, the current findings will help improve disease control in shrimps.
Collapse
Affiliation(s)
- Chuanqi Wang
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Lingwei Ruan
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Hong Shi
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Wenyang Lin
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China.,School of Life Science, Xiamen University, Xiamen, China
| | - Linmin Liu
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| | - Sujie Li
- State Key Laboratory Breeding Base of Marine Genetic Resources, Key Laboratory of Marine Genetic Resources of Ministry of Natural Resources, Third Institute of Oceanography, Ministry of Natural Resources, Fujian Key Laboratory of Marine Genetic Resources, Xiamen, China
| |
Collapse
|
28
|
Zhang Y, Wu J, Zeng C, Xu L, Wei W, Li Y. The role of NFAT2/miR-20a-5p signaling pathway in the regulation of CD8 + naïve T cells activation and differentiation. Immunobiology 2021; 226:152111. [PMID: 34237654 DOI: 10.1016/j.imbio.2021.152111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022]
Abstract
T cell dysfunction is a common characteristic in leukemia patients that significantly impacts clinical treatment and prognosis. However, the mechanism underlying T cell dysfunction and its reversal remains unclear. In this study, in accordance with our previous findings, we found that the expression of NFAT2 and pri-miR-17 ~ 92 are lower in peripheral blood CD3+ T cells from chronic myelogenous leukemia (CML) patients by gene expression analysis. We further demonstrate that the NFAT2-induced activation, differentiation, and expression of cytokines in human umbilical cord blood CD8+ naïve T cells are miR-20a-5p dependent. We also preliminarily explored the relationship between NFAT2 and miR-20a-5p in naive T cells. These results suggest that NFAT2 and miR-20a are crucial for regulating functional CD8+ T cells. Additionally, their alteration may be related to CD8+ T cell dysfunction in CML patients; thus, NFAT2 and miR-20a-5p may be considered potential targets for revising T cell function in leukemia immunotherapy.
Collapse
Affiliation(s)
- Yikai Zhang
- Depart of Hematology, First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou 510663, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, China
| | - Jialu Wu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, China
| | - Chengwu Zeng
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, China
| | - Ling Xu
- Depart of Hematology, First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, China.
| | - Wei Wei
- Guangzhou Municipality Tianhe Nuoya Bio-engineering Co. Ltd, Guangzhou 510663, China.
| | - Yangqiu Li
- Depart of Hematology, First Affiliated Hospital, Jinan University, Guangzhou 510632, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, 601 Huang Pu Da Dao Xi, 510632 Guangzhou, China.
| |
Collapse
|
29
|
Rothenberg EV. Logic and lineage impacts on functional transcription factor deployment for T-cell fate commitment. Biophys J 2021; 120:4162-4181. [PMID: 33838137 PMCID: PMC8516641 DOI: 10.1016/j.bpj.2021.04.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/22/2021] [Accepted: 04/02/2021] [Indexed: 11/19/2022] Open
Abstract
Transcription factors are the major agents that read the regulatory sequence information in the genome to initiate changes in expression of specific genes, both in development and in physiological activation responses. Their actions depend on site-specific DNA binding and are largely guided by their individual DNA target sequence specificities. However, their action is far more conditional in a real developmental context than would be expected for simple reading of local genomic DNA sequence, which is common to all cells in the organism. They are constrained by slow-changing chromatin states and by interactions with other transcription factors, which affect their occupancy patterns of potential sites across the genome. These mechanisms lead to emergent discontinuities in function even for transcription factors with minimally changing expression. This is well revealed by diverse lineages of blood cells developing throughout life from hematopoietic stem cells, which use overlapping combinations of transcription factors to drive strongly divergent gene regulation programs. Here, using development of T lymphocytes from hematopoietic multipotent progenitor cells as a focus, recent evidence is reviewed on how binding specificity and dynamics, transcription factor cooperativity, and chromatin state changes impact the effective regulatory functions of key transcription factors including PU.1, Runx1, Notch-RBPJ, and Bcl11b.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California.
| |
Collapse
|
30
|
Fernando N, Sciumè G, O'Shea JJ, Shih HY. Multi-Dimensional Gene Regulation in Innate and Adaptive Lymphocytes: A View From Regulomes. Front Immunol 2021; 12:655590. [PMID: 33841440 PMCID: PMC8034253 DOI: 10.3389/fimmu.2021.655590] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/09/2021] [Indexed: 12/24/2022] Open
Abstract
The precise control of cytokine production by innate lymphoid cells (ILCs) and their T cell adaptive system counterparts is critical to mounting a proper host defense immune response without inducing collateral damage and autoimmunity. Unlike T cells that differentiate into functionally divergent subsets upon antigen recognition, ILCs are developmentally programmed to rapidly respond to environmental signals in a polarized manner, without the need of T cell receptor (TCR) signaling. The specification of cytokine production relies on dynamic regulation of cis-regulatory elements that involve multi-dimensional epigenetic mechanisms, including DNA methylation, transcription factor binding, histone modification and DNA-DNA interactions that form chromatin loops. How these different layers of gene regulation coordinate with each other to fine tune cytokine production, and whether ILCs and their T cell analogs utilize the same regulatory strategy, remain largely unknown. Herein, we review the molecular mechanisms that underlie cell identity and functionality of helper T cells and ILCs, focusing on networks of transcription factors and cis-regulatory elements. We discuss how higher-order chromatin architecture orchestrates these components to construct lineage- and state-specific regulomes that support ordered immunoregulation.
Collapse
Affiliation(s)
- Nilisha Fernando
- Neuro-Immune Regulome Unit, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Giuseppe Sciumè
- Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci-Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - John J O'Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Han-Yu Shih
- Neuro-Immune Regulome Unit, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.,National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
31
|
Rothenberg EV. Single-cell insights into the hematopoietic generation of T-lymphocyte precursors in mouse and human. Exp Hematol 2021; 95:1-12. [PMID: 33454362 PMCID: PMC8018899 DOI: 10.1016/j.exphem.2020.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 01/29/2023]
Abstract
T-Cell development is a major branch of lymphoid development and a key output of hematopoiesis, especially in early life, but the molecular requirements for T-cell potential have remained obscure. Considerable advances have now been made toward solving this problem through single-cell transcriptome studies, interfaced with in vitro differentiation assays that monitor potential efficiently at the single-cell level. This review focuses on a series of recent reports studying mouse and human early T-cell precursors, both in the developing fetus and in stringently purified postnatal samples of intrathymic and prethymic T-lineage precursors. Cross-comparison of results reveals a robustly conserved core program in mouse and human, but with some informative and provocative variations between species and between ontogenic states. Repeated findings are the multipotent progenitor regulatory signature of thymus-seeding cells and the proximity of the T-cell program to dendritic cell programs, especially to plasmacytoid dendritic cells in humans.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA.
| |
Collapse
|
32
|
Hosokawa H, Rothenberg EV. How transcription factors drive choice of the T cell fate. Nat Rev Immunol 2021; 21:162-176. [PMID: 32918063 PMCID: PMC7933071 DOI: 10.1038/s41577-020-00426-6] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2020] [Indexed: 12/21/2022]
Abstract
Recent evidence has elucidated how multipotent blood progenitors transform their identities in the thymus and undergo commitment to become T cells. Together with environmental signals, a core group of transcription factors have essential roles in this process by directly activating and repressing specific genes. Many of these transcription factors also function in later T cell development, but control different genes. Here, we review how these transcription factors work to change the activities of specific genomic loci during early intrathymic development to establish T cell lineage identity. We introduce the key regulators and highlight newly emergent insights into the rules that govern their actions. Whole-genome deep sequencing-based analysis has revealed unexpectedly rich relationships between inherited epigenetic states, transcription factor-DNA binding affinity thresholds and influences of given transcription factors on the activities of other factors in the same cells. Together, these mechanisms determine T cell identity and make the lineage choice irreversible.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
33
|
Elsaid R, Meunier S, Burlen-Defranoux O, Soares-da-Silva F, Perchet T, Iturri L, Freyer L, Vieira P, Pereira P, Golub R, Bandeira A, Perdiguero EG, Cumano A. A wave of bipotent T/ILC-restricted progenitors shapes the embryonic thymus microenvironment in a time-dependent manner. Blood 2021; 137:1024-1036. [PMID: 33025012 PMCID: PMC8065239 DOI: 10.1182/blood.2020006779] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
During embryonic development, multiple waves of hematopoietic progenitors with distinct lineage potential are differentially regulated in time and space. Two different waves of thymic progenitors colonize the fetal thymus where they contribute to thymic organogenesis and homeostasis. The origin, the lineage differentiation potential of the first wave, and their relative contribution in shaping the thymus architecture, remained, however, unclear. Here, we show that the first wave of thymic progenitors comprises a unique population of bipotent T and innatel lymphoid cells (T/ILC), generating a lymphoid tissue inducer cells (LTi's), in addition to invariant Vγ5+ T cells. Transcriptional analysis revealed that innate lymphoid gene signatures and, more precisely, the LTi-associated transcripts were expressed in the first, but not in the second, wave of thymic progenitors. Depletion of early thymic progenitors in a temporally controlled manner showed that the progeny of the first wave is indispensable for the differentiation of autoimmune regulator-expressing medullary thymic epithelial cells (mTECs). We further show that these progenitors are of strict hematopoietic stem cell origin, despite the overlap between lymphopoiesis initiation and the transient expression of lymphoid-associated transcripts in yolk sac (YS) erythromyeloid-restricted precursors. Our work highlights the relevance of the developmental timing on the emergence of different lymphoid subsets, required for the establishment of a functionally diverse immune system.
Collapse
Affiliation(s)
- Ramy Elsaid
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sylvain Meunier
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Odile Burlen-Defranoux
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Francisca Soares-da-Silva
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Instituto de Investigação e Inovação em Saúde (I3S) and
- Instituto Nacional de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Thibaut Perchet
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Lorea Iturri
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
- Cellule Pasteur, University Pierre et Marie Curie (UPMC), Paris, France
| | - Laina Freyer
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
| | - Paulo Vieira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pablo Pereira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Rachel Golub
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Antonio Bandeira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Elisa Gomez Perdiguero
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
| | - Ana Cumano
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
34
|
Hosokawa H, Masuhara K, Koizumi M. Transcription factors regulate early T cell development via redeployment of other factors: Functional dynamics of constitutively required factors in cell fate decisions. Bioessays 2021; 43:e2000345. [PMID: 33624856 DOI: 10.1002/bies.202000345] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 01/02/2023]
Abstract
Establishment of cell lineage identity from multipotent progenitors is controlled by cooperative actions of lineage-specific and stably expressed transcription factors, combined with input from environmental signals. Lineage-specific master transcription factors activate and repress gene expression by recruiting consistently expressed transcription factors and chromatin modifiers to their target loci. Recent technical advances in genome-wide and multi-omics analysis have shed light on unexpected mechanisms that underlie more complicated actions of transcription factors in cell fate decisions. In this review, we discuss functional dynamics of stably expressed and continuously required factors, Notch and Runx family members, throughout developmental stages of early T cell development in the thymus. Pre- and post-commitment stage-specific transcription factors induce dynamic redeployment of Notch and Runx binding genomic regions. Thus, together with stage-specific transcription factors, shared transcription factors across distinct developmental stages regulate acquisition of T lineage identity.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan.,Institute of Medical Sciences, Tokai University, Isehara, Kanagawa, Japan
| | - Kaori Masuhara
- Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Maria Koizumi
- Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| |
Collapse
|
35
|
Liu C, Lan Y, Liu B, Zhang H, Hu H. T Cell Development: Old Tales Retold By Single-Cell RNA Sequencing. Trends Immunol 2021; 42:165-175. [PMID: 33446417 DOI: 10.1016/j.it.2020.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Mammalian T cell development initiates from the migration of hematopoietic progenitors to the thymus, which undergo cell proliferation, T-lineage specification and commitment, as well as positive and negative selection. These processes are precisely controlled at multiple levels and have been intensively studied using gene-modified animal models and in vitro coculture systems. However, several long-standing questions, including the characterization of the rare but crucial progenitors/precursors and the molecular mechanisms underlying their fate decision, have been dampened because of cell scarcity and lack of appropriate techniques. Single-cell RNA sequencing (scRNA-seq) makes it possible to investigate and resolve some of these questions, leading to new remarkable progress in identifying and characterizing early thymic progenitors and delineating the refined developmental trajectories of conventional and unconventional T cells.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China; State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Huiyuan Zhang
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Hongbo Hu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
36
|
Olariu V, Yui MA, Krupinski P, Zhou W, Deichmann J, Andersson E, Rothenberg EV, Peterson C. Multi-scale Dynamical Modeling of T Cell Development from an Early Thymic Progenitor State to Lineage Commitment. Cell Rep 2021; 34:108622. [PMID: 33440162 DOI: 10.1016/j.celrep.2020.108622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 04/24/2020] [Accepted: 12/18/2020] [Indexed: 01/13/2023] Open
Abstract
Intrathymic development of committed progenitor (pro)-T cells from multipotent hematopoietic precursors offers an opportunity to dissect the molecular circuitry establishing cell identity in response to environmental signals. This transition encompasses programmed shutoff of stem/progenitor genes, upregulation of T cell specification genes, proliferation, and ultimately commitment. To explain these features in light of reported cis-acting chromatin effects and experimental kinetic data, we develop a three-level dynamic model of commitment based upon regulation of the commitment-linked gene Bcl11b. The levels are (1) a core gene regulatory network (GRN) architecture from transcription factor (TF) perturbation data, (2) a stochastically controlled chromatin-state gate, and (3) a single-cell proliferation model validated by experimental clonal growth and commitment kinetic assays. Using RNA fluorescence in situ hybridization (FISH) measurements of genes encoding key TFs and measured bulk population dynamics, this single-cell model predicts state-switching kinetics validated by measured clonal proliferation and commitment times. The resulting multi-scale model provides a mechanistic framework for dissecting commitment dynamics.
Collapse
Affiliation(s)
- Victor Olariu
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Mary A Yui
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pawel Krupinski
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Wen Zhou
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA
| | - Julia Deichmann
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Emil Andersson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Carsten Peterson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, Sweden.
| |
Collapse
|
37
|
Liu Q, Kim MH, Friesen L, Kim CH. BATF regulates innate lymphoid cell hematopoiesis and homeostasis. Sci Immunol 2020; 5:eaaz8154. [PMID: 33277375 PMCID: PMC8375455 DOI: 10.1126/sciimmunol.aaz8154] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 06/01/2020] [Accepted: 11/09/2020] [Indexed: 12/31/2022]
Abstract
Early hematopoietic progenitors undergo sophisticated developmental processes to become committed innate lymphoid cell (ILC) progenitors and ultimately mature ILC subsets in the periphery. Basic leucine zipper ATF-like transcription factor (Batf) plays important roles in lymphocyte biology. We report here that Batf regulates the production of bone marrow ILC progenitors and maintenance of peripheral ILCs. The expression of Batf is induced during ILC development at the α-lymphoid progenitor stage in response to the cytokine IL-7. As a potential mechanism, up-regulated Batf binds and activates transcription of the Nfil3 gene to promote ILC hematopoiesis. Batf is necessary to maintain normal numbers of early and late ILC progenitors in the bone marrow and mature ILC1, ILC2, ILC3, and NK cells in most peripheral tissues. Batf deficiency causes ILC lymphopenia, leading to defective ILC responses to inflammatory cytokines and defective immunity to enteric bacterial infections. Thus, Batf plays critical roles in bone marrow hematopoiesis, peripheral homeostasis, and effector functions of ILCs.
Collapse
Affiliation(s)
- Qingyang Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Myung H Kim
- Laboratory of Immunology and Hematopoiesis, Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Leon Friesen
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
- Mary H. Weiser Food Allergy Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| |
Collapse
|
38
|
Qian K, Xu JX, Deng Y, Peng H, Peng J, Ou CM, Liu Z, Jiang LH, Tai YH. Signaling pathways of genetic variants and miRNAs in the pathogenesis of myasthenia gravis. Gland Surg 2020; 9:1933-1944. [PMID: 33447544 PMCID: PMC7804555 DOI: 10.21037/gs-20-39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 09/30/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Myasthenia gravis (MG) is a chronic autoimmune neuromuscular disorder causing muscle weakness and characterized by a defect in synaptic transmission at the neuromuscular junction. The pathogenesis of this disease remains unclear. We aimed to predict the key signaling pathways of genetic variants and miRNAs in the pathogenesis of MG, and identify the key genes among them. METHODS We searched published information regarding associated single nucleotide polymorphisms (SNPs) and differentially-expressed miRNAs in MG cases. We search of SNPs and miRNAs in literature databases about MG, then we used bioinformatic tools to predict target genes of miRNAs. Moreover, functional enrichment analysis for key genes was carried out utilizing the Cytoscape-plugin, known as ClueGO. These key genes were mapped to STRING database to construct a protein-protein interaction (PPI) network. Then a miRNA-target gene regulatory network was established to screen key genes. RESULTS Five genes containing SNPs associated with MG risk were involved in the inflammatory bowel disease (IBD) signaling pathway, and FoxP3 was the key gene. MAPK1, SMAD4, SMAD2 and BCL2 were predicted to be targeted by the 18 miRNAs and to act as the key genes in adherens, junctions, apoptosis, or cancer-related pathways respectively. These five key genes containing SNPs or targeted by miRNAs were found to be involved in negative regulation of T cell differentiation. CONCLUSIONS We speculate that SNPs cause the genes to be defective or the miRNAs to downregulate the factors that subsequently negatively regulate regulatory T cells and trigger the onset of MG.
Collapse
Affiliation(s)
- Kai Qian
- Faculty of Life and Biotechnology, Kunming University of Science and Technology, Kunming, China
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jia-Xin Xu
- Department of Cardiovascular surgery, Yan’ an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi Deng
- Department of Oncology, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Peng
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Jun Peng
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Chun-Mei Ou
- Department of Cardiovascular surgery, Institute of the First People’s Hospital of Yunnan Province, Kunming, China
| | - Zu Liu
- Department of Cardiovascular surgery, Yan’ an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li-Hong Jiang
- Department of Thoracic Surgery, Institute of The First People’s Hospital of Yunnan Province, Kunming, China
| | - Yong-Hang Tai
- School of Electronic Information in the Yunnan Normal University, Kunming, China
| |
Collapse
|
39
|
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: 1.4] [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
|
40
|
Yu H, Hu W, Song X, Zhao Y. Notch-HEY2 signaling pathway contributes to the differentiation of CD34 + hematopoietic-like stem cells from adult peripheral blood insulin-producing cells after the treatment with platelet-derived mitochondria. Mol Biol Rep 2020; 47:8347-8352. [PMID: 32997309 DOI: 10.1007/s11033-020-05874-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/28/2020] [Indexed: 11/30/2022]
Abstract
Previous works characterized a novel cell population from adult human peripheral blood, designated peripheral blood insulin-producing cells (PB-IPC). PB-IPC displayed the pluripotent potential of differentiations after the treatment with platelet-derived mitochondria and gave rise to three germ layer-derived cells such as the mitochondrion-induced CD34+ hematopoietic stem cells (HSC)-like cells (miCD34+ HSC). To determine the molecular mechanism underlying the differentiation of miCD34+ cells, mechanistic studies established that MitoTracker Deep Red-labeled mitochondria could enter into the PB-IPC in a dose-dependent manner. Blocking Notch signaling pathway with a γ-secretase inhibitor, DAPT, markedly inhibited the proliferation of PB-IPC and improved the differentiation of miCD34+ HSC. Additionally, treatment with platelet-derived mitochondria can reprogram the differentiation of PB-IPC into miCD34+ HSC through inhibition of the Notch/HEY2 signaling pathway, as demonstrated by blocking experiments with HEY2 small interfering RNA (siRNA). The data indicated that Notch signaling pathway contributes to the miCD34+ HSC differentiation, thus advancing our understanding of the mitochondrial reprogramming and the potential treatment of human hematopoietic disease.
Collapse
Affiliation(s)
- Haibo Yu
- Center for Discovery and Innovation, Hackensack Meridian Health, 340 Kingsland Street, Nutley, NJ, 07110, USA
| | - Wei Hu
- Center for Discovery and Innovation, Hackensack Meridian Health, 340 Kingsland Street, Nutley, NJ, 07110, USA
| | - Xiang Song
- Center for Discovery and Innovation, Hackensack Meridian Health, 340 Kingsland Street, Nutley, NJ, 07110, USA
| | - Yong Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, 340 Kingsland Street, Nutley, NJ, 07110, USA.
| |
Collapse
|
41
|
Hosokawa H, Romero-Wolf M, Yang Q, Motomura Y, Levanon D, Groner Y, Moro K, Tanaka T, Rothenberg EV. Cell type-specific actions of Bcl11b in early T-lineage and group 2 innate lymphoid cells. J Exp Med 2020; 217:jem.20190972. [PMID: 31653691 PMCID: PMC7037248 DOI: 10.1084/jem.20190972] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/16/2019] [Accepted: 09/27/2019] [Indexed: 01/16/2023] Open
Abstract
Bcl11b binds to distinctive genomic regions with different partners and regulates completely different target genes in pro-T and ILC2 cells. Despite these divergences in Bcl11b function, a shared enhancer supports initial Bcl11b locus opening in both pro-T and ILC2 lineages. The zinc finger transcription factor, Bcl11b, is expressed in T cells and group 2 innate lymphoid cells (ILC2s) among hematopoietic cells. In early T-lineage cells, Bcl11b directly binds and represses the gene encoding the E protein antagonist, Id2, preventing pro-T cells from adopting innate-like fates. In contrast, ILC2s co-express both Bcl11b and Id2. To address this contradiction, we have directly compared Bcl11b action mechanisms in pro-T cells and ILC2s. We found that Bcl11b binding to regions across the genome shows distinct cell type–specific motif preferences. Bcl11b occupies functionally different sites in lineage-specific patterns and controls totally different sets of target genes in these cell types. In addition, Bcl11b bears cell type–specific post-translational modifications and organizes different cell type–specific protein complexes. However, both cell types use the same distal enhancer region to control timing of Bcl11b activation. Therefore, although pro-T cells and ILC2s both need Bcl11b for optimal development and function, Bcl11b works substantially differently in these two cell types.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA.,Department of Immunology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Maile Romero-Wolf
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Qi Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY
| | - Yasutaka Motomura
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Laboratory for Innate Immune Systems, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomoaki Tanaka
- Department of Molecular Diagnosis, Graduate School of Medicine, Chiba University, Chiba, Japan.,Agency for Medical Research and Development - Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
42
|
Cumano A, Berthault C, Ramond C, Petit M, Golub R, Bandeira A, Pereira P. New Molecular Insights into Immune Cell Development. Annu Rev Immunol 2020; 37:497-519. [PMID: 31026413 DOI: 10.1146/annurev-immunol-042718-041319] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During development innate lymphoid cells and specialized lymphocyte subsets colonize peripheral tissues, where they contribute to organogenesis and later constitute the first line of protection while maintaining tissue homeostasis. A few of these subsets are produced only during embryonic development and remain in the tissues throughout life. They are generated through a unique developmental program initiated in lympho-myeloid-primed progenitors, which lose myeloid and B cell potential. They either differentiate into innate lymphoid cells or migrate to the thymus to give rise to embryonic T cell receptor-invariant T cells. At later developmental stages, adaptive T lymphocytes are derived from lympho-myeloid progenitors that colonize the thymus, while lymphoid progenitors become specialized in the production of B cells. This sequence of events highlights the requirement for stratification in the establishment of immune functions that determine efficient seeding of peripheral tissues by a limited number of cells.
Collapse
Affiliation(s)
- Ana Cumano
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Claire Berthault
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Cyrille Ramond
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , ,
| | - Maxime Petit
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Rachel Golub
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Antonio Bandeira
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Pablo Pereira
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| |
Collapse
|
43
|
Generation of Hematopoietic-Like Stem Cells from Adult Human Peripheral Blood Following Treatment with Platelet-Derived Mitochondria. Int J Mol Sci 2020; 21:ijms21124249. [PMID: 32549211 PMCID: PMC7352808 DOI: 10.3390/ijms21124249] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/06/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Adult stem cells represent a potential source for cellular therapy to treat serious human diseases. We characterized the insulin-producing cells from adult peripheral blood (designated PB-IPC), which displayed a unique phenotype. Mitochondria are normally located in the cellular cytoplasm, where they generate ATP to power the cell’s functions. Ex vivo and in vivo functional studies established that treatment with platelet-derived mitochondria can reprogram the transformation of adult PB-IPC into functional CD34+ hematopoietic stem cells (HSC)-like cells, leading to the production of blood cells such as T cells, B cells, monocytes/macrophages, granulocytes, red blood cells, and megakaryocytes (MKs)/platelets. These findings revealed a novel function of mitochondria in directly contributing to cellular reprogramming, thus overcoming the limitations and safety concerns of using conventional technologies to reprogram embryonic stem (ES) and induced pluripotent stem (iPS) cells in regenerative medicine.
Collapse
|
44
|
Dolens A, Durinck K, Lavaert M, Van der Meulen J, Velghe I, De Medts J, Weening K, Roels J, De Mulder K, Volders P, De Preter K, Kerre T, Vandekerckhove B, Leclercq G, Vandesompele J, Mestdagh P, Van Vlierberghe P, Speleman F, Taghon T. Distinct Notch1 and BCL11B requirements mediate human γδ/αβ T cell development. EMBO Rep 2020; 21:e49006. [PMID: 32255245 PMCID: PMC7202205 DOI: 10.15252/embr.201949006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 03/03/2020] [Accepted: 03/12/2020] [Indexed: 12/22/2022] Open
Abstract
γδ and αβ T cells have unique roles in immunity and both originate in the thymus from T-lineage committed precursors through distinct but unclear mechanisms. Here, we show that Notch1 activation is more stringently required for human γδ development compared to αβ-lineage differentiation and performed paired mRNA and miRNA profiling across 11 discrete developmental stages of human T cell development in an effort to identify the potential Notch1 downstream mechanism. Our data suggest that the miR-17-92 cluster is a Notch1 target in immature thymocytes and that miR-17 can restrict BCL11B expression in these Notch-dependent T cell precursors. We show that enforced miR-17 expression promotes human γδ T cell development and, consistently, that BCL11B is absolutely required for αβ but less for γδ T cell development. This study suggests that human γδ T cell development is mediated by a stage-specific Notch-driven negative feedback loop through which miR-17 temporally restricts BCL11B expression and provides functional insights into the developmental role of the disease-associated genes BCL11B and the miR-17-92 cluster in a human context.
Collapse
Affiliation(s)
| | - Kaat Durinck
- Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Marieke Lavaert
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | | | - Imke Velghe
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Jelle De Medts
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Karin Weening
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Juliette Roels
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
- Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | | | | | | | - Tessa Kerre
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | | | | | - Jo Vandesompele
- Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Pieter Mestdagh
- Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | | | - Frank Speleman
- Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Tom Taghon
- Department of Diagnostic SciencesGhent UniversityGhentBelgium
| |
Collapse
|
45
|
Miyazato K, Hayakawa Y. Pharmacological targeting of natural killer cells for cancer immunotherapy. Cancer Sci 2020; 111:1869-1875. [PMID: 32301190 PMCID: PMC7293096 DOI: 10.1111/cas.14418] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/07/2020] [Accepted: 04/10/2020] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that rapidly respond to cancer cells without prior sensitization or restriction to the cognate antigen in comparison with tumor antigen-specific T cells. Recent advances in understanding NK-cell biology have elucidated the molecular mechanisms underlying the differentiation and maturation of NK cells, in addition to the control of their effector functions by investigating the receptors and ligands involved in the recognition of cancer cells by NK cells. Such clarification of NK-cell recognition of cancer cells also revealed the mechanism by which cancer cells potentially evade NK-cell-dependent immune surveillance. Furthermore, the recent clinical results of T-cell-targeted cancer immunotherapy have increased the expectations for new immunotherapies by targeting NK cells. However, the potential use of NK cells in cancer immunotherapy is not fully understood. In this review, we discuss the current evidence and future potential of pharmacological targeting of NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Kiho Miyazato
- Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | | |
Collapse
|
46
|
Rao TN, Kumar S, Pulikkottil AJ, Oliveri F, Hendriks RW, Beckel F, Fehling HJ. Novel, Non-Gene-Destructive Knock-In Reporter Mice Refute the Concept of Monoallelic Gata3 Expression. THE JOURNAL OF IMMUNOLOGY 2020; 204:2600-2611. [PMID: 32213568 DOI: 10.4049/jimmunol.2000025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/26/2020] [Indexed: 02/04/2023]
Abstract
Accurately tuned expression levels of the transcription factor GATA-3 are crucial at several stages of T cell and innate lymphoid cell development and differentiation. Moreover, several lines of evidence suggest that Gata3 expression might provide a reliable molecular marker for the identification of elusive progenitor cell subsets at the earliest stages of T lineage commitment. To be able to faithfully monitor Gata3 expression noninvasively at the single-cell level, we have generated a novel strain of knock-in reporter mice, termed GATIR, by inserting an expression cassette encoding a bright fluorescent marker into the 3'-untranslated region of the endogenous Gata3 locus. Importantly, in contrast to three previously published strains of Gata3 reporter mice, GATIR mice preserve physiological Gata3 expression on the targeted allele. In this study, we show that GATIR mice faithfully reflect endogenous Gata3 expression without disturbing the development of GATA-3-dependent lymphoid cell populations. We further show that GATIR mice provide an ideal tool for noninvasive monitoring of Th2 polarization and straightforward identification of innate lymphoid cell 2 progenitor populations. Finally, as our reporter is non-gene-destructive, GATIR mice can be bred to homozygosity, not feasible with previously published strains of Gata3 reporter mice harboring disrupted alleles. The availability of hetero- and homozygous Gata3 reporter mice with an exceptionally bright fluorescent marker, allowed us to visualize allelic Gata3 expression in individual cells simply by flow cytometry. The unambiguous results obtained provide compelling evidence against previously postulated monoallelic Gata3 expression in early T lineage and hematopoietic stem cell subsets.
Collapse
Affiliation(s)
| | - Suresh Kumar
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | | | - Franziska Oliveri
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus Medical Center, NL-3000 CA Rotterdam, the Netherlands
| | - Franziska Beckel
- Institute of Immunology, University Hospital, D-89081 Ulm, Germany; and
| | | |
Collapse
|
47
|
Lavaert M, Liang KL, Vandamme N, Park JE, Roels J, Kowalczyk MS, Li B, Ashenberg O, Tabaka M, Dionne D, Tickle TL, Slyper M, Rozenblatt-Rosen O, Vandekerckhove B, Leclercq G, Regev A, Van Vlierberghe P, Guilliams M, Teichmann SA, Saeys Y, Taghon T. Integrated scRNA-Seq Identifies Human Postnatal Thymus Seeding Progenitors and Regulatory Dynamics of Differentiating Immature Thymocytes. Immunity 2020; 52:1088-1104.e6. [PMID: 32304633 DOI: 10.1016/j.immuni.2020.03.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/04/2020] [Accepted: 03/27/2020] [Indexed: 10/24/2022]
Abstract
During postnatal life, thymopoiesis depends on the continuous colonization of the thymus by bone-marrow-derived hematopoietic progenitors that migrate through the bloodstream. The current understanding of the nature of thymic immigrants is largely based on data from pre-clinical models. Here, we employed single-cell RNA sequencing (scRNA-seq) to examine the immature postnatal thymocyte population in humans. Integration of bone marrow and peripheral blood precursor datasets identified two putative thymus seeding progenitors that varied in expression of CD7; CD10; and the homing receptors CCR7, CCR9, and ITGB7. Whereas both precursors supported T cell development, only one contributed to intrathymic dendritic cell (DC) differentiation, predominantly of plasmacytoid dendritic cells. Trajectory inference delineated the transcriptional dynamics underlying early human T lineage development, enabling prediction of transcription factor (TF) modules that drive stage-specific steps of human T cell development. This comprehensive dataset defines the expression signature of immature human thymocytes and provides a resource for the further study of human thymopoiesis.
Collapse
Affiliation(s)
- Marieke Lavaert
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium
| | - Kai Ling Liang
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium
| | - Niels Vandamme
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jong-Eun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Juliette Roels
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Monica S Kowalczyk
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Bo Li
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Sciences Platform, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Marcin Tabaka
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Timothy L Tickle
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Haematology Department, Royal Victoria Infirmary, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Bart Vandekerckhove
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Howard Hughes Medical Institute, Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Pieter Van Vlierberghe
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Ontogeny and Functional Specialization, VIB Center for Inflammation Research, Ghent, Belgium; Faculty of Sciences, Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK; Theory of Condensed Matter Group, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Tom Taghon
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, C. Heymanslaan 10, MRB2, Entrance 38, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.
| |
Collapse
|
48
|
Assessing Phosphorylation of STAT Transcription Factors in Mouse Innate Lymphoid Cells. Methods Mol Biol 2020. [PMID: 32147786 DOI: 10.1007/978-1-0716-0338-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Innate lymphoid cells (ILCs) ensure protection against pathogens by quickly reacting to the alterations of the cytokine milieu taking place upon infection. More than 50 cytokines and growth factors activate the Janus kinases (JAKs), leading to phosphorylation of members of the signal transducer and activator of transcription (STAT) family. Activation of STATs induces specific transcriptional programs which are associated with distinct cellular outcomes. Thus, an efficient measurement of rapid STAT phosphorylation enables not only to dissect the spectrum of cytokine sensitivity among ILC subsets but also to pinpoint specific transcriptional programs and cellular functions initiated after activation. Using this method, we have previously dissected the downstream events of Interleukin (IL)-23 and IL-12 signaling in ILCs, shedding light on the differential usage of STATs among ILC subsets. Here, we provide an optimized and detailed protocol describing how to analyze phosphorylation of STAT transcription factors in murine NK and ILC subsets isolated from different tissues.
Collapse
|
49
|
Ghaedi M, Shen ZY, Orangi M, Martinez-Gonzalez I, Wei L, Lu X, Das A, Heravi-Moussavi A, Marra MA, Bhandoola A, Takei F. Single-cell analysis of RORα tracer mouse lung reveals ILC progenitors and effector ILC2 subsets. J Exp Med 2020; 217:e20182293. [PMID: 31816636 PMCID: PMC7062532 DOI: 10.1084/jem.20182293] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/12/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022] Open
Abstract
Lung group 2 innate lymphoid cells (ILC2s) drive allergic inflammation and promote tissue repair. ILC2 development is dependent on the transcription factor retinoic acid receptor-related orphan receptor (RORα), which is also expressed in common ILC progenitors. To elucidate the developmental pathways of lung ILC2s, we generated RORα lineage tracer mice and performed single-cell RNA sequencing, flow cytometry, and functional analyses. In adult mouse lungs, we found an IL-18Rα+ST2- population different from conventional IL-18Rα-ST2+ ILC2s. The former was GATA-3intTcf7EGFP+Kit+, produced few cytokines, and differentiated into multiple ILC lineages in vivo and in vitro. In neonatal mouse lungs, three ILC populations were identified, namely an ILC progenitor population similar to that in adult lungs and two distinct effector ILC2 subsets that differentially produced type 2 cytokines and amphiregulin. Lung ILC progenitors might actively contribute to ILC-poiesis in neonatal and inflamed adult lungs. In addition, neonatal lung ILC2s include distinct proinflammatory and tissue-repairing subsets.
Collapse
Affiliation(s)
- Maryam Ghaedi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Zi Yi Shen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Mona Orangi
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, British Columbia, Canada
| | - Itziar Martinez-Gonzalez
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| | - Lisa Wei
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Xiaoxiao Lu
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alireza Heravi-Moussavi
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Fumio Takei
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada
| |
Collapse
|
50
|
Marcel N, Hedrick SM. A key control point in the T cell response to chronic infection and neoplasia: FOXO1. Curr Opin Immunol 2020; 63:51-60. [PMID: 32135399 DOI: 10.1016/j.coi.2020.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 01/29/2020] [Accepted: 02/01/2020] [Indexed: 12/26/2022]
Abstract
T cells able to control neoplasia or chronic infections display a signature gene expression profile similar or identical to that of central memory T cells. These cells have qualities of self-renewal and a plasticity that allow them to repeatedly undergo activation (growth, proliferation, and differentiation), followed by quiescence. It is these qualities that define the ability of T cells to establish an equilibrium with chronic infectious agents, and also preserve the ability of T cells to be re-activated (by checkpoint therapy) in response to malignant cancers. Here we describe distinctions between the forms of inhibition mediated by tumors and persistent viruses, we review the properties of T cells associated with long-term immunity, and we identify the transcription factor, FOXO1, as the control point for a program of gene expression that allows CD8+ T cells to undergo serial reactivation and self-renewal.
Collapse
Affiliation(s)
- Nimi Marcel
- Molecular Biology Section, Division of Biological Sciences, Department of Cellular and Molecular Medicine, TATA Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093-0377, United States
| | - Stephen M Hedrick
- Molecular Biology Section, Division of Biological Sciences, Department of Cellular and Molecular Medicine, TATA Institute for Genetics and Society, University of California, San Diego, La Jolla, CA 92093-0377, United States.
| |
Collapse
|