1
|
Blümke J, Schameitat M, Verma A, Limbecker C, Arlt E, Kessler SM, Kielstein H, Krug S, Bazwinsky-Wutschke I, Haemmerle M. Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer. Cancers (Basel) 2025; 17:1689. [PMID: 40427186 PMCID: PMC12110028 DOI: 10.3390/cancers17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar-ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Juliane Blümke
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| | - Moritz Schameitat
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Atul Verma
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
| | - Celina Limbecker
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sonja M. Kessler
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Faculty of Natural Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany;
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Sebastian Krug
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (A.V.); (S.K.)
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany; (M.S.); (C.L.); (H.K.); (I.B.-W.)
| | - Monika Haemmerle
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany;
| |
Collapse
|
2
|
Li J, Wang H, Xia S. Hematopoietic stem and progenitor cells fine-tuning the "sweet" of trained immunity. J Leukoc Biol 2025; 117:qiaf043. [PMID: 40233187 DOI: 10.1093/jleuko/qiaf043] [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: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Recent studies have challenged the traditional view of innate immunity as nonspecific and transient by demonstrating that innate immune cells can develop immune memory in response to various activating factors, a phenomenon known as trained immunity. This process involves epigenetic modifications, such as changes in chromatin accessibility, and metabolic reprogramming, which can provide protection against unrelated pathogens but may also trigger immune-mediated damage. This review summarizes the current understanding of innate immune memory, with a particular focus on recent findings regarding the training of innate immune cells at the hematopoietic stem and progenitor cell stage. We present observations of trained immunity in innate immune cells, summarize key activating factors and underlying mechanisms, and propose potential host-directed immunotherapeutic strategies and preventive measures based on trained immunity. Our aim is to highlight the biological significance of trained immunity and its potential applications in enhancing long-term immunity, improving vaccine efficacy, and preventing immune-related diseases.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| |
Collapse
|
3
|
Byatt TC, Razaghi E, Tüzüner S, Simões FC. Immune-mediated cardiac development and regeneration. Semin Cell Dev Biol 2025; 171:103613. [PMID: 40315634 DOI: 10.1016/j.semcdb.2025.103613] [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: 12/29/2024] [Revised: 03/18/2025] [Accepted: 04/16/2025] [Indexed: 05/04/2025]
Abstract
The complex interplay between the immune and cardiovascular systems during development, homeostasis and regeneration represents a rapidly evolving field in cardiac biology. Single cell technologies, spatial mapping and computational analysis have revolutionised our understanding of the diversity and functional specialisation of immune cells within the heart. From the earliest stages of cardiogenesis, where primitive macrophages guide heart tube formation, to the complex choreography of inflammation and its resolution during regeneration, immune cells emerge as central orchestrators of cardiac fate. Translating these fundamental insights into clinical applications represents a major challenge and opportunity for the field. In this Review, we decode the immunological blueprint of heart development and regeneration to transform cardiovascular disease treatment and unlock the regenerative capacity of the human heart.
Collapse
Affiliation(s)
- Timothy C Byatt
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Ehsan Razaghi
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Selin Tüzüner
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Filipa C Simões
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
4
|
Galstyan DS, Kolesnikova TO, Demin KA, Dubrovskii YA, Murashko E, Kessenikh E, Ilyin NP, Ikrin AN, Moskalenko AM, de Abreu MS, Yang L, Kalueff AV. Intranasal delivery of drugs to the central nervous system of adult zebrafish. Lab Anim (NY) 2025; 54:126-132. [PMID: 40307351 DOI: 10.1038/s41684-025-01545-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/18/2025] [Indexed: 05/02/2025]
Abstract
The small teleost zebrafish (Danio rerio) has become a critically important laboratory animal in biomedicine. One of their key practical advantages, the convenient method of small-molecule administration via water immersion, has certain problems with dosing precision and drug delivery. Here, we present a simple protocol for the intranasal delivery of neuroactive drugs in adult zebrafish using arecoline and nicotine, two well-studied reference neuroactive drugs chosen for the proof of concept. Adult fish received 1 μL water solution of arecoline (1 and 10 mg/mL) or nicotine tartrate (5 and 10 mg/mL) or the same volume of drug-free water (control) into both nostrils, followed by behavioral testing in the novel tank test 5 min later. Mass spectrometry analyses confirmed that both drugs rapidly reached the zebrafish brain following intranasal administration. Intranasally administered arecoline (10 mg/mL) and nicotine (5 and 10 mg/mL) demonstrated overt behavioral profiles, evoking characteristic anxiolytic-like effects in zebrafish similar to those observed here for a standard 20-min water immersion method (10 mg/L arecoline or 30 mg/L nicotine). Overall, we showed that neuroactive drugs can be delivered to adult zebrafish intranasally to exert central effects, which may (at least for some drugs) occur faster and can need smaller drug quantities than for the water immersion delivery.
Collapse
Affiliation(s)
- David S Galstyan
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana O Kolesnikova
- Neuroscience Department, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Yaroslav A Dubrovskii
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Ekaterina Murashko
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Elizaveta Kessenikh
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Nikita P Ilyin
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Aleksey N Ikrin
- Neuroscience Department, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Anastasia M Moskalenko
- Neuroscience Department, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil.
- Western Caspian University, Baku, Azerbaijan.
| | - Longen Yang
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Suzhou Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Allan V Kalueff
- World Class Research Center for Personalized Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.
- Neuroscience Department, Sirius University of Science and Technology, Sirius Federal Territory, Russia.
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
- Suzhou Key Laboratory of Neurobiology and Cell Signaling, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China.
| |
Collapse
|
5
|
Ghani F, Zubair AC. Possible impacts of cosmic radiation on leukemia development during human deep space exploration. Leukemia 2025:10.1038/s41375-025-02624-4. [PMID: 40275072 DOI: 10.1038/s41375-025-02624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/14/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025]
Abstract
With the advent of deep space exploration and ambitious plans to return humans to the Moon and journey onward to Mars, humans will face exposure to ionizing radiation beyond Earth's atmosphere and magnetosphere. This is particularly concerning for the hematopoietic system that is sensitive to galactic cosmic rays (GCRs) during interplanetary missions. Epidemiological studies and animal studies implicate that exposure to ionizing radiation can cause leukemias, with recent consensus showing that almost all types of leukemias, even chronic lymphocytic leukemia, can be caused by ionizing radiation despite previous controversies. The possible deleterious effects of deep space travel on the formation, development, etiology, and pathophysiology of hematologic malignancies, specifically leukemias, remain largely unclear. The mechanism(s) by which ionizing radiations cause leukemia differs for different leukemia types and is poorly understood in the spaceflight environment, posing a serious health risk for future astronauts. This paper provides a comprehensive review of the various studies and evidence available on Earth and in space assessing the relationship between ionizing radiation and increased risk of leukemia. We also discuss the unique characteristics of leukemia in space, ethical considerations, risk assessments and potential challenges this may bring to astronauts and healthcare professionals as humanity continues to explore the cosmos.
Collapse
Affiliation(s)
- Fay Ghani
- Center for Regenerative Biotherapeutics and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - Abba C Zubair
- Center for Regenerative Biotherapeutics and Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
6
|
Yang W, Peng M, Wang Y, Zhang X, Li W, Zhai X, Wu Z, Hu P, Chen L. Deletion of hepcidin disrupts iron homeostasis and hematopoiesis in zebrafish embryogenesis. Development 2025; 152:dev204307. [PMID: 40110772 DOI: 10.1242/dev.204307] [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: 08/02/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
Iron is essential for cell growth and hematopoiesis, which is regulated by hepcidin (hamp). However, the role of hamp in zebrafish hematopoiesis remains unclear. Here, we have created a stable hamp knockout zebrafish model using the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated nuclease 9 system (CRISPR/Cas9 system). Our study revealed that hamp deletion led to maternal iron overload in embryos, significantly downregulating hemoglobin genes and reducing hemoglobin content. Single-cell RNA sequencing identified abnormal expression patterns in blood progenitor cells, with a specific progenitor subtype showing increased ferroptosis and delayed development. By crossing hamp knockout zebrafish with a gata1+ line (blood cells labeled fish line), we confirmed ferroptosis in blood progenitor cells. These findings underscore the crucial role of hamp in iron regulation and hematopoiesis, offering novel insights into developmental biology and potential therapeutic targets for blood disorders.
Collapse
Affiliation(s)
- Wenyi Yang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Mingjian Peng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Youquan Wang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Xiaowen Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wei Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Xue Zhai
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zhichao Wu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Liangbiao Chen
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China
- International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
7
|
Nordin A, Zambanini G, Enar Jonasson M, Weiss T, van de Grift Y, Pagella P, Cantù C. Construction of an atlas of transcription factor binding during mouse development identifies popular regulatory regions. Development 2025; 152:dev204311. [PMID: 40013513 DOI: 10.1242/dev.204311] [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: 08/06/2024] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Gene regulators physically associate with the genome, in a combinatorial fashion, to drive tissue-specific gene expression. Uncovering the genome-wide activity of all gene regulators across tissues is therefore needed to understand gene regulation during development. Here, we take a first step towards this goal. Using CUT&RUN, we systematically mapped genome-wide binding profiles of key transcription factors and co-factors that mediate ontogenetically relevant signaling pathways in select mouse tissues at two developmental stages. Computation of the datasets unveiled tissue- and time-specific activity for each gene regulator. We identified 'popular' regulatory regions that are bound by a multitude of regulators, which tend to be more evolutionarily conserved. Consistently, they lie near the transcription start site of genes for which dysregulation results in early embryonic lethality. Moreover, the human homologs of these regions are similarly bound by many gene regulators and are highly conserved, indicating a retained relevance for human development. This work constitutes a decisive step towards understanding how the genome is simultaneously read and used by gene regulators in a holistic fashion to drive embryonic development.
Collapse
Affiliation(s)
- Anna Nordin
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
- Science for Life Laboratory - SciLifeLab, Linköping University, 58185 Linköping, Sweden
| | - Gianluca Zambanini
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
| | - Mattias Enar Jonasson
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
| | - Tamina Weiss
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
- Science for Life Laboratory - SciLifeLab, Linköping University, 58185 Linköping, Sweden
| | - Yorick van de Grift
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
- Science for Life Laboratory - SciLifeLab, Linköping University, 58185 Linköping, Sweden
| | - Pierfrancesco Pagella
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
| | - Claudio Cantù
- Wallenberg Centre for Molecular Medicine, Linköping University, 58185 Linköping, Sweden
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology, Faculty of Medicine and Health Sciences, Linköping University, 58185 Linköping, Sweden
- Science for Life Laboratory - SciLifeLab, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
8
|
Lin G, Wang Y, Pham TG, Wen Z. Dendritic cells in developing and adult zebrafish arise from different origins and display distinct flt3 dependencies. Development 2025; 152:DEV204410. [PMID: 39866089 DOI: 10.1242/dev.204410] [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: 09/23/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025]
Abstract
Dendritic cells (DCs) are key cellular components of the immune system and perform crucial functions in innate and acquired immunity. In mammals, it is generally believed that DCs originate exclusively from hematopoietic stem cells (HSCs). Using a temporal-spatial resolved fate-mapping system, here we show that, in zebrafish, DCs arise from two sources: dorsal aorta-born endothelium-derived hematopoietic progenitors (EHPs) and HSCs. The EHP-derived DCs emerge early, predominantly colonizing the developing thymus during larval stages and diminishing by juvenile stages. In contrast, HSC-derived DCs emerge later and can populate different tissues from late larval stages to adulthood. We further document that the EHP- and HSC-derived DCs display different dependencies on Fms-like tyrosine kinase 3 (Flt3), a pivotal receptor tyrosine kinase crucial for DC development in mammals. Our study reveals the presence of two distinct waves of DC development in zebrafish, each with unique origins and developmental controls.
Collapse
Affiliation(s)
- Guanzhen Lin
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 000000, China
| | - Youqi Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 000000, China
| | - Thi Giang Pham
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong 000000, China
| | - Zilong Wen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
9
|
Liu X, Sha J, Wang L, Wang Z, Fang Z, Han X, Tan S, Chen Y, Yuan H, De The H, Zhou J, Zhu J. Rnf111 has a pivotal role in regulating development of definitive hematopoietic stem and progenitor cells through the Smad2/3-Gcsfr/NO axis in zebrafish. Haematologica 2025; 110:385-396. [PMID: 39363867 PMCID: PMC11788642 DOI: 10.3324/haematol.2024.285438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
The ubiquitination or SUMOylation of hematopoietic-related factors plays pivotal roles in hematopoiesis. RNF111, known as a ubiquitin ligase, is a newly discovered SUMO-targeted ubiquitin ligase involved in multiple signaling pathways mediated by transforming growth factor (TGF)-β family members. However, its role in hematopoiesis remains unclear. Herein, a heritable Rnf111 mutant zebrafish line was generated by CRISPR/Cas9-mediated genome editing. Impairment of hematopoietic stem and progenitor cells (HSPC) of definitive hematopoiesis was found in Rnf111-deficient mutants. Ablation of Rnf111 resulted in decreased phosphorylation of Smad2/3 in HSPC. Definitive endoderm 2 inducer (IDE2), which specifically activates TGF-β signaling and downstream Smad2 phosphorylation, could restore definitive hematopoiesis in Rnf111-deficient embryos. Further molecular mechanism studies revealed that Gcsfr/NO signaling was an important target pathway of Smad2/3 involved in Rnf111-mediated HSPC development. In conclusion, our study demonstrated that Rnf111 contributes to the development of HSPC by maintaining Smad2/3 phosphorylation and activation of the Gcsfr/NO signaling pathway.
Collapse
Affiliation(s)
- Xiaohui Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025.
| | - Jinghan Sha
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Luxiang Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Zixuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Zhou Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Xiao Han
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Shuiyi Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Yi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Hao Yuan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Hugues De The
- CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris 75010
| | - Jun Zhou
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025
| | - Jun Zhu
- CNRS IRP (International research Project), Cancer, Aging and Hematology, Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Université de Paris 7/INSERM/CNRS UMR 944/7212, Equipe Labellisée Ligue Nationale Contre le Cancer, Hôpital St. Louis, Paris 75010.
| |
Collapse
|
10
|
Woelk J, Narasimhan H, Pfeifhofer-Obermair C, Schraml BU, Hermann-Kleiter N. NR2F6 regulates stem cell hematopoiesis and myelopoiesis in mice. Front Immunol 2025; 15:1404805. [PMID: 39840064 PMCID: PMC11747239 DOI: 10.3389/fimmu.2024.1404805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Nuclear receptors regulate hematopoietic stem cells (HSCs) and peripheral immune cells in mice and humans. The nuclear orphan receptor NR2F6 (EAR-2) has been shown to control murine hematopoiesis. Still, detailed analysis of the distinct stem cell, myeloid, and lymphoid progenitors in the bone marrow in a genetic loss of function model remains pending. In this study, we found that adult germline Nr2f6-deficient mice contained increased percentages of total long-term and short-term HSCs, as well as a subpopulation within the lineage-biased multipotent progenitor (MPP3) cells. The loss of NR2F6 thus led to an increase in the percentage of LSK+ cells. Following the differentiation from the common myeloid progenitors (CMP), the granulocyte-monocyte progenitors (GMP) were decreased, while monocyte-dendritic progenitors (MDP) were increased in Nr2f6-deficient bone marrow. Within the pre-conventional dendritic progenitors (pre-cDCs), the subpopulation of pre-cDC2s was reduced in the bone marrow of Nr2f6-deficient mice. We did not observe differences in the development of common lymphoid progenitor populations. Our findings contrast previous studies but underscore the role of NR2F6 in regulating gene expression levels during mouse bone marrow hematopoiesis and myelopoiesis.
Collapse
Affiliation(s)
- Johannes Woelk
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hamsa Narasimhan
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology at the Walter-Brendel-Centre of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christa Pfeifhofer-Obermair
- Department of Internal Medicine II (Infectious Diseases, Immunology, Rheumatology, Pneumology), Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara U. Schraml
- Institute for Immunology, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology at the Walter-Brendel-Centre of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
11
|
Liu S, Feng Z, Su M, Liu C, Xi Y, Chen H, Luo L, Tian X, Zhao F, Li L. Tango6 regulates HSPC proliferation and definitive haematopoiesis via Ikzf1 and Cmyb in caudal haematopoietic tissue. Development 2025; 152:dev202903. [PMID: 39620979 DOI: 10.1242/dev.202903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/30/2024] [Indexed: 01/04/2025]
Abstract
Haematopoietic stem and progenitor cells (HSPCs) arise from the aorta-gonad-mesonephros and migrate to the caudal haematopoietic tissue (CHT) in zebrafish, where nascent HSPCs undergo tightly controlled proliferation and differentiation to promote definitive haematopoiesis. Effective expansion of HSPCs requires the coordination of well-established vesicle trafficking systems and appropriate transcription factors. However, the underlying molecules are yet to be identified. Using large-scale genetic screening of zebrafish larvae, Tango6 of the coat protein complex I (COPI) vesicle trafficking system was found to be indispensable for HSPC proliferation and definitive haematopoiesis. Homozygous tango6cq72 mutants display defective expansion of HSPCs in the CHT and compromised haematopoiesis. However, haematopoietic overexpression of Tango6 promoted haematopoietic expansion. tango6 deficiency caused a decline in RNA polymerase II subunit B and accumulation of DNA damage, which suppressed cell expansion in a P53-dependent manner. ikzf1 and cmyb (myb), two indispensable haematopoietic transcription factors, are targets of P53 and are used by tango6 in haematopoiesis. The haematopoietic phenotype was partially recovered by compensating for loss of ikzf1 and cmyb in tango6cq72 mutants. This study reveals a vesicle trafficking-mediated Tango6-P53-Ikzf1/Cmyb axis in zebrafish definitive haematopoiesis.
Collapse
Affiliation(s)
- Shengnan Liu
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Zhi Feng
- Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Ming Su
- Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Chenchen Liu
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Yuan Xi
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Huan Chen
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Fangying Zhao
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, Chongqing 400715, China
| | - Li Li
- Research Center of Stem cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| |
Collapse
|
12
|
van der Weele CM, Hospes KC, Rowe KE, Jeffery WR. Hypoxia-sonic hedgehog axis as a driver of primitive hematopoiesis development and evolution in cavefish. Dev Biol 2024; 516:138-147. [PMID: 39173434 PMCID: PMC11402556 DOI: 10.1016/j.ydbio.2024.08.008] [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: 06/11/2024] [Revised: 08/09/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
The teleost Astyanax mexicanus consists of surface dwelling (surface fish) and cave dwelling (cavefish) forms. Cavefish have evolved in subterranean habitats characterized by reduced oxygen levels (hypoxia) and exhibit a subset of phenotypic traits controlled by increased Sonic hedgehog (Shh) signaling along the embryonic midline. The enhancement of primitive hematopoietic domains, which are formed bilaterally in the anterior and posterior lateral plate mesoderm, are responsible for the development of more larval erythrocytes in cavefish relative to surface fish. In this study, we determine the role of hypoxia and Shh signaling in the development and evolution of primitive hematopoiesis in cavefish. We show that hypoxia treatment during embryogenesis increases primitive hematopoiesis and erythrocyte development in surface fish. We also demonstrate that upregulation of Shh midline signaling by the Smoothened agonist SAG increases primitive hematopoiesis and erythrocyte development in surface fish, whereas Shh downregulation via treatment with the Smoothened inhibitor cyclopamine decreases these traits in cavefish. Together these results suggest that hematopoietic enhancement is regulated by hypoxia and Shh signaling. Lastly, we demonstrate that hypoxia enhances expression of Shh signaling along the midline of surface fish embryos. We conclude that hypoxia-mediated Shh plasticity may be a driving force for the adaptive evolution of primitive hematopoiesis and erythrocyte development in cavefish.
Collapse
Affiliation(s)
| | - Katrina C Hospes
- Department of Biology, University of Maryland, College Park, MD, 20742, USA
| | - Katherine E Rowe
- Department of Biology, University of Maryland, College Park, MD, 20742, USA
| | - William R Jeffery
- Department of Biology, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
13
|
Shi LL, Ye K, Wang SZ, Hou CJ, Song AK, Liu H, Wang HL. Deletion of the foxO1 gene reduces hypoxia tolerance in zebrafish embryos by influencing erythropoiesis. Life Sci 2024; 357:123048. [PMID: 39270834 DOI: 10.1016/j.lfs.2024.123048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/24/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
FoxO1 (Forkhead box O1) belongs to the evolutionarily conserved FoxO subfamily and is involved in diverse physiologic processes, including apoptosis, cell cycle, DNA damage repair, oxidative stress and cell differentiation. FoxO1 plays an important role in regulating the hypoxia microenvironment such as cancers, but its role in hypoxia adaptation remains unclear in animals. To understand the function of foxO1 in hypoxia response, we constructed foxO1a and foxO1b mutant zebrafish using CRISPR/Cas9 technology. It was found that foxO1a and foxO1b destruction affected the hematopoietic system in the early zebrafish embryos. Specifically, FoxO1a and FoxO1b were found to affect the transcriptional activity of runx1, a marker gene for hematopoietic stem cells (HSCs). Moreover, foxO1a and foxO1b had complementary features in hypoxia response, and foxO1a or/and foxO1b destruction resulted in tolerance of zebrafish becoming weakened in hypoxia due to insufficient hemoglobin supply. Additionally, the transcriptional activity of these two genes was demonstrated to be regulated by Hif1α. In conclusion, foxO1a and foxO1b respond to Hif1α-mediated hypoxia response by participating in zebrafish erythropoiesis. These results will provide a theoretical basis for further exploring the function of FoxO1 in hematopoiesis and hypoxia response.
Collapse
Affiliation(s)
- Lin-Lin Shi
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Ke Ye
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Su-Zhen Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Chao-Jie Hou
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - An-Kang Song
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Hong Liu
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China
| | - Huan-Ling Wang
- Key Lab of Freshwater Animal Breeding, Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, 430070 Wuhan, PR China.
| |
Collapse
|
14
|
Franza M, Varricchio R, Alloisio G, De Simone G, Di Bella S, Ascenzi P, di Masi A. Zebrafish ( Danio rerio) as a Model System to Investigate the Role of the Innate Immune Response in Human Infectious Diseases. Int J Mol Sci 2024; 25:12008. [PMID: 39596075 PMCID: PMC11593600 DOI: 10.3390/ijms252212008] [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: 10/07/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
The zebrafish (Danio rerio) has emerged as a valuable model for studying host-pathogen interactions due to its unique combination of characteristics. These include extensive sequence and functional conservation with the human genome, optical transparency in larvae that allows for high-resolution visualization of host cell-microbe interactions, a fully sequenced and annotated genome, advanced forward and reverse genetic tools, and suitability for chemical screening studies. Despite anatomical differences with humans, the zebrafish model has proven instrumental in investigating immune responses and human infectious diseases. Notably, zebrafish larvae rely exclusively on innate immune responses during the early stages of development, as the adaptive immune system becomes fully functional only after 4-6 weeks post-fertilization. This window provides a unique opportunity to isolate and examine infection and inflammation mechanisms driven by the innate immune response without the confounding effects of adaptive immunity. In this review, we highlight the strengths and limitations of using zebrafish as a powerful vertebrate model to study innate immune responses in infectious diseases. We will particularly focus on host-pathogen interactions in human infections caused by various bacteria (Clostridioides difficile, Staphylococcus aureus, and Pseudomonas aeruginosa), viruses (herpes simplex virus 1, SARS-CoV-2), and fungi (Aspergillus fumigatus and Candida albicans).
Collapse
Affiliation(s)
- Maria Franza
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Romualdo Varricchio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giulia Alloisio
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Giovanna De Simone
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
| | - Stefano Di Bella
- Clinical Department of Medical, Surgical and Health Sciences, Trieste University, 34127 Trieste, Italy;
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Accademia Nazionale dei Lincei, 00165 Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, 00146 Roma, Italy; (M.F.); (R.V.); (G.A.); (G.D.S.); (P.A.)
- Centro Linceo Interdisciplinare “Beniamino Segre”, Accademia Nazionale dei Lincei, 00165 Roma, Italy
| |
Collapse
|
15
|
Chen K, Wu J, Zhang Y, Liu W, Chen X, Zhang W, Huang Z. Cebpa is required for haematopoietic stem and progenitor cell generation and maintenance in zebrafish. Open Biol 2024; 14:240215. [PMID: 39500381 PMCID: PMC11537755 DOI: 10.1098/rsob.240215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 11/09/2024] Open
Abstract
The CCAAT enhancer binding protein alpha (CEBPA) is crucial for myeloid differentiation and the balance of haematopoietic stem and progenitor cell (HSPC) quiescence and self-renewal, and its dysfunction can drive leukemogenesis. However, its role in HSPC generation has not been fully elucidated. Here, we utilized various zebrafish cebpa mutants to investigate the function of Cebpa in the HSPC compartment. Co-localization analysis showed that cebpa expression is enriched in nascent HSPCs. Complete loss of Cebpa function resulted in a significant reduction in early HSPC generation and the overall HSPC pool during embryonic haematopoiesis. Interestingly, while myeloid differentiation was impaired in cebpa N-terminal mutants expressing the truncated zP30 protein, the number of HSPCs was not affected, indicating a redundant role of Cebpa P42 and P30 isoforms in HSPC development. Additionally, epistasis experiments confirmed that Cebpa functions downstream of Runx1 to regulate HSPC emergence. Our findings uncover a novel role of Cebpa isoforms in HSPC generation and maintenance, and provide new insights into HSPC development.
Collapse
Affiliation(s)
- Kemin Chen
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| | - Jieyi Wu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| | - Yuxian Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| | - Wei Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| | - Xiaohui Chen
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| | - Wenqing Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong518055, People’s Republic of China
| | - Zhibin Huang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, Guangdong510006, People’s Republic of China
| |
Collapse
|
16
|
Kharrat B, Gábor E, Virág N, Sinka R, Jankovics F, Kristó I, Vilmos P, Csordás G, Honti V. Dual role for Headcase in hemocyte progenitor fate determination in Drosophila melanogaster. PLoS Genet 2024; 20:e1011448. [PMID: 39466810 PMCID: PMC11515969 DOI: 10.1371/journal.pgen.1011448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
The hematopoietic organ of the Drosophila larva, the lymph gland, is a simplified representation of mammalian hematopoietic compartments, with the presence of hemocyte progenitors in the medullary zone (MZ), differentiated hemocytes in the cortical zone (CZ), and a hematopoietic niche called the posterior signaling centre (PSC) that orchestrates progenitor differentiation. Our previous work has demonstrated that the imaginal cell factor Headcase (Hdc, Heca) is required in the hematopoietic niche to control the differentiation of hemocyte progenitors. However, the downstream mechanisms of Hdc-mediated hematopoietic control remained unknown. Here we show that Hdc exerts this function by negatively regulating the insulin/mTOR signaling in the niche. When Hdc is depleted in the PSC, the overactivation of this pathway triggers reactive oxygen species (ROS) accumulation and, in turn, the differentiation of effector lamellocytes non-cell-autonomously. Although overactivation of insulin/mTOR signaling normally leads to an increase in the size of the hematopoietic niche, this effect is concealed by cell death caused by hdc loss-of-function. Moreover, we describe here that hdc silencing in progenitors causes cell-autonomous ROS elevation and JNK pathway activation, resulting in decreased MZ size and differentiation of lamellocytes. Similarly to the PSC niche, knocking down hdc in the MZ also leads to caspase activation. Notably, depleting Hdc in the progenitors triggers proliferation, an opposing effect to what is observed in the niche. These findings further our understanding of how progenitor maintenance in the larval lymph gland is controlled autonomously and non-cell-autonomously, and point towards new mechanisms potentially regulating HSC maintenance across vertebrates.
Collapse
Affiliation(s)
- Bayan Kharrat
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Erika Gábor
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Nikolett Virág
- Department of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Rita Sinka
- Department of Genetics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Ferenc Jankovics
- Laboratory of Drosophila Germ Cell Differentiation, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Ildikó Kristó
- Drosophila Nuclear Actin Laboratory, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Péter Vilmos
- Drosophila Nuclear Actin Laboratory, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Gábor Csordás
- Lysosomal Degradation Research Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Viktor Honti
- Drosophila Blood Cell Differentiation Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| |
Collapse
|
17
|
Ling Y, Wu J, Liu Y, Meng P, Sun Y, Zhao D, Lin Q. Establishment of a Diamond-Blackfan anemia like model in zebrafish. Dev Dyn 2024; 253:906-921. [PMID: 38450920 DOI: 10.1002/dvdy.703] [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: 07/25/2023] [Revised: 02/03/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Anemia is defined as a lack of erythrocytes, low hemoglobin levels, or abnormal erythrocyte morphology. Diamond-Blackfan anemia (DBA) is a rare and severe congenital hypoplastic anemia that occurs due to the dominant inheritance of a ribosomal protein gene mutation. Even rarer is a case described as Diamond-Blackfan anemia like (DBAL), which occurs due to a loss-of-function EPO mutation recessive inheritance. The effective cures for DBAL are bone marrow transfusion and treatment with erythropoiesis-stimulating agents (ESAs). To effectively manage the condition, construction of DBAL models to identify new medical methods or screen drugs are necessary. RESULTS Here, an epoa-deficient mutant zebrafish called epoaszy8 was generated to model DBAL. The epoa-deficiency in zebrafish caused developmental defects in erythroid cells, leading to severe congenital anemia. Using the DBAL model, we validated a loss-of-function EPO mutation using an in vivo functional analysis and explored the ability of ESAs to alleviate congenital anemia. CONCLUSIONS Together, our study demonstrated that epoa deficiency in zebrafish leads to a phenotype resembling DBAL. The DBAL zebrafish model was found to be beneficial for the in vivo assessment of patient-derived EPO variants with unclear implications and for devising potential therapeutic approaches for DBAL.
Collapse
Affiliation(s)
- Yiming Ling
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jiaye Wu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yushi Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Panpan Meng
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ying Sun
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| | - Dejian Zhao
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Qing Lin
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
18
|
Gunage R, Zon LI. Role of RNA modifications in blood development and regeneration. Exp Hematol 2024; 138:104279. [PMID: 39009277 DOI: 10.1016/j.exphem.2024.104279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/17/2024]
Abstract
Blood development and regeneration require rapid turnover of cells, and ribonucleic acid (RNA) modifications play a key role in it via regulating stemness and cell fate regulation. RNA modifications affect gene activity via posttranscriptional and translation-mediated mechanisms. Diverse molecular players involved in RNA-modification processes are abundantly expressed by hematopoietic stem cells and lineages. Close to 150 RNA chemical modifications have been reported, but only N6-methyl adenosine (m6A), inosine (I), pseudouridine (Ψ), and m1A-a handful-have been studied in-cell fate regulation. The role of RNA modification in blood diseases and disorders is an emerging field and offers potential for therapeutic interventions. Knowledge of RNA-modification and enzymatic activities could be used to design therapies in the future. Here, we summarized the recent advances in RNA modification and the epitranscriptome field and discussed their regulation of blood development and regeneration.
Collapse
Affiliation(s)
- Rajesh Gunage
- Stem Cell Program and Division of Hematology/Oncology, Department of Medicine, Children's Hospital Boston, Harvard Stem Cell Institute, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Department of Medicine, Children's Hospital Boston, Harvard Stem Cell Institute, Harvard Medical School and Howard Hughes Medical Institute, Boston, MA.
| |
Collapse
|
19
|
So KWL, Su Z, Cheung JPY, Choi SW. Single-Cell Analysis of Bone-Marrow-Disseminated Tumour Cells. Diagnostics (Basel) 2024; 14:2172. [PMID: 39410576 PMCID: PMC11475990 DOI: 10.3390/diagnostics14192172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Metastasis frequently targets bones, where cancer cells from the primary tumour migrate to the bone marrow, initiating new tumour growth. Not only is bone the most common site for metastasis, but it also often marks the first site of metastatic recurrence. Despite causing over 90% of cancer-related deaths, effective treatments for bone metastasis are lacking, with current approaches mainly focusing on palliative care. Circulating tumour cells (CTCs) are pivotal in metastasis, originating from primary tumours and circulating in the bloodstream. They facilitate metastasis through molecular interactions with the bone marrow environment, involving direct cell-to-cell contacts and signalling molecules. CTCs infiltrate the bone marrow, transforming into disseminated tumour cells (DTCs). While some DTCs remain dormant, others become activated, leading to metastatic growth. The presence of DTCs in the bone marrow strongly correlates with future bone and visceral metastases. Research on CTCs in peripheral blood has shed light on their release mechanisms, yet investigations into bone marrow DTCs have been limited. Challenges include the invasiveness of bone marrow aspiration and the rarity of DTCs, complicating their isolation. However, advancements in single-cell analysis have facilitated insights into these elusive cells. This review will summarize recent advancements in understanding bone marrow DTCs using single-cell analysis techniques.
Collapse
Affiliation(s)
| | | | | | - Siu-Wai Choi
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (K.W.L.S.); (Z.S.); (J.P.Y.C.)
| |
Collapse
|
20
|
Kanbay M, Mutlu A, Bakir CN, Peltek IB, Canbaz AA, Díaz Tocados JM, Haarhaus M. Klotho in pregnancy and intrauterine development-potential clinical implications: a review from the European Renal Association CKD-MBD Working Group. Nephrol Dial Transplant 2024; 39:1574-1582. [PMID: 38486352 PMCID: PMC11427066 DOI: 10.1093/ndt/gfae066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Indexed: 09/28/2024] Open
Abstract
Intrauterine development is crucial for life-long health; therefore, elucidation of its key regulators is of interest for their potential prognostic and therapeutic implications. Originally described as a membrane-bound anti-aging protein, Klotho has evolved as a regulator of numerous functions in different organ systems. Circulating Klotho is generated by alternative splicing or active shedding from cell membranes. Recently, Klotho was identified as a regulator of placental function, and while Klotho does not cross the placental barrier, increased levels of circulating α-Klotho have been identified in umbilical cord blood compared with maternal blood, indicating that Klotho may also play a role in intrauterine development. In this narrative review, we discuss novel insights into the specific functions of the Klotho proteins in the placenta and in intrauterine development, while summarizing up-to-date knowledge about their structures and functions. Klotho plays a role in stem cell functioning, organogenesis and haematopoiesis. Low circulating maternal and foetal levels of Klotho are associated with preeclampsia, intrauterine growth restriction, and an increased perinatal risk for newborns, indicating a potential use of Klotho as biomarker and therapeutic target. Experimental administration of Klotho protein indicates a neuro- and nephroprotective potential, suggesting a possible future role of Klotho as a therapeutic agent. However, the use of Klotho as intervention during pregnancy is as yet unproven. Here, we summarize novel evidence, suggesting Klotho as a key regulator for healthy pregnancies and intrauterine development with promising potential for clinical use.
Collapse
Affiliation(s)
- Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Ali Mutlu
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Cicek N Bakir
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ibrahim B Peltek
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Ata A Canbaz
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Juan Miguel Díaz Tocados
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida, Dr Pifarré Foundation (IRBLleida), Lleida, Spain
| | - Mathias Haarhaus
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
- Diaverum AB, Malmö, Sweden
| |
Collapse
|
21
|
Lints R, Walker CA, Delfi O, Prouse M, PohLui De Silva M, Bohlander SK, Wood AC. Mutational cooperativity of RUNX1::RUNX1T1 isoform 9a and oncogenic NRAS in zebrafish myeloid leukaemia. Biol Open 2024; 13:bio060523. [PMID: 39177514 PMCID: PMC11381922 DOI: 10.1242/bio.060523] [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: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
RUNX1::RUNX1T1 (R::RT1) acute myeloid leukaemia (AML) remains a clinical challenge, and further research is required to model and understand leukaemogenesis. Previous zebrafish R::RT1 models were hampered by embryonic lethality and low penetrance of the malignant phenotype. Here, we overcome this by developing an adult zebrafish model in which the human R::RT1 isoform 9a is co-expressed with the frequently co-occurring oncogenic NRASG12D mutation in haematopoietic stem and progenitor cells (HSPCs), using the Runx1+23 enhancer. Approximately 50% of F0 9a+NRASG12D transgenic zebrafish developed signs of haematological disease between 5 and 14 months, with 27% exhibiting AML-like pathology: myeloid precursor expansion, erythrocyte reduction, kidney marrow hypercellularity and the presence of blasts. Moreover, only 9a+NRASG12D transplant recipients developed leukaemia with high rates of mortality within 40 days, inferring the presence of leukaemia stem cells. These leukaemic features were rare or not observed in animals expressing either the NRAS or 9a oncogenes alone, suggesting 9a and NRAS cooperation drives leukaemogenesis. This novel adult AML zebrafish model provides a powerful new tool for investigating the basis of R::RT1 - NRAS cooperativity with the potential to uncover new therapeutic targets.
Collapse
Affiliation(s)
- Robyn Lints
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Christina A. Walker
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Omid Delfi
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Matthew Prouse
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | | | - Stefan K. Bohlander
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
| | - Andrew C. Wood
- Leukaemia and Blood Cancer Research Unit, Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1023, New Zealand
- Starship Child Health, Starship Blood and Cancer Centre, Auckland 1023, New Zealand
| |
Collapse
|
22
|
Khattab S, El Sorady M, El-Ghandour A, Visani G, Piccaluga PP. Hematopoietic and leukemic stem cells homeostasis: the role of bone marrow niche. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1027-1055. [PMID: 39351440 PMCID: PMC11438561 DOI: 10.37349/etat.2024.00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/01/2024] [Indexed: 10/04/2024] Open
Abstract
The bone marrow microenvironment (BMM) has highly specialized anatomical characteristics that provide a sanctuary place for hematopoietic stem cells (HSCs) that allow appropriate proliferation, maintenance, and self-renewal capacity. Several cell types contribute to the constitution and function of the bone marrow niche. Interestingly, uncovering the secrets of BMM and its interaction with HSCs in health paved the road for research aiming at better understanding the concept of leukemic stem cells (LSCs) and their altered niche. In fact, they share many signals that are responsible for interactions between LSCs and the bone marrow niche, due to several biological similarities between LSCs and HSCs. On the other hand, LSCs differ from HSCs in their abnormal activation of important signaling pathways that regulate survival, proliferation, drug resistance, invasion, and spread. Targeting these altered niches can help in better treatment choices for hematological malignancies and bone marrow disorders in general and acute myeloid leukemia (AML) in particular. Moreover, targeting those niches may help in decreasing the emergence of drug resistance and lower the relapse rate. In this article, the authors reviewed the most recent literature on bone marrow niches and their relations with either normal HSCs and AML cells/LSC, by focusing on pathogenetic and therapeutic implications.
Collapse
Affiliation(s)
- Shaimaa Khattab
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
- Medical Research Institute, Hematology department, Alexandria University, Alexandria 21561, Egypt
| | - Manal El Sorady
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Ashraf El-Ghandour
- Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria 5310002, Egypt
| | - Giuseppe Visani
- Hematology and Stem Cell Transplant Center, Azienda Ospedaliera Marche Nord, 61121 Pesaro, Italy
| | - Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Ospedaliera-Universitaria di Bologna Policlinico di S. Orsola, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, Bologna University School of Medicine, 40138 Bologna, Italy
| |
Collapse
|
23
|
Doll L, Welte K, Skokowa J, Bajoghli B. A JAGN1-associated severe congenital neutropenia zebrafish model revealed an altered G-CSFR signaling and UPR activation. Blood Adv 2024; 8:4050-4065. [PMID: 38739706 PMCID: PMC11342096 DOI: 10.1182/bloodadvances.2023011656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024] Open
Abstract
ABSTRACT A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.
Collapse
Affiliation(s)
- Larissa Doll
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Karl Welte
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Children’s Hospital, University Hospital Tuebingen, Tuebingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Gene and RNA Therapy Center, Tuebingen University, Tuebingen, Germany
| | - Baubak Bajoghli
- Department of Oncology, Hematology, Clinical Immunology, and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
- Austrian BioImaging/CMI, Vienna, Austria
| |
Collapse
|
24
|
Quarato ER, Salama NA, Calvi LM. Interplay Between Skeletal and Hematopoietic Cells in the Bone Marrow Microenvironment in Homeostasis and Aging. Curr Osteoporos Rep 2024; 22:416-432. [PMID: 38782850 DOI: 10.1007/s11914-024-00874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE OF THE REVIEW In this review, we discuss the most recent scientific advances on the reciprocal regulatory interactions between the skeletal and hematopoietic stem cell niche, focusing on immunomodulation and its interplay with the cell's mitochondrial function, and how this impacts osteoimmune health during aging and disease. RECENT FINDINGS Osteoimmunology investigates interactions between cells that make up the skeletal stem cell niche and immune system. Much work has investigated the complexity of the bone marrow microenvironment with respect to the skeletal and hematopoietic stem cells that regulate skeletal formation and immune health respectively. It has now become clear that these cellular components cooperate to maintain homeostasis and that dysfunction in their interaction can lead to aging and disease. Having a deeper, mechanistic appreciation for osteoimmune regulation will lead to better research perspective and therapeutics with the potential to improve the aging process, skeletal and hematologic regeneration, and disease targeting.
Collapse
Affiliation(s)
- Emily R Quarato
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| | - Noah A Salama
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
| | - Laura M Calvi
- James P. Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
25
|
Phng LK, Hogan BM. Endothelial cell transitions in zebrafish vascular development. Dev Growth Differ 2024; 66:357-368. [PMID: 39072708 PMCID: PMC11457512 DOI: 10.1111/dgd.12938] [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: 05/07/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024]
Abstract
In recent decades, developmental biologists have come to view vascular development as a series of progressive transitions. Mesoderm differentiates into endothelial cells; arteries, veins and lymphatic endothelial cells are specified from early endothelial cells; and vascular networks diversify and invade developing tissues and organs. Our understanding of this elaborate developmental process has benefitted from detailed studies using the zebrafish as a model system. Here, we review a number of key developmental transitions that occur in zebrafish during the formation of the blood and lymphatic vessel networks.
Collapse
Affiliation(s)
- Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Benjamin M Hogan
- Organogenesis and Cancer Programme, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology and the Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
26
|
Sukhnanan K, Ross JR, Chao NJ, Chen BJ. Endothelial Cell Derived Extracellular Vesicles and Hematopoiesis. Radiat Res 2024; 202:215-226. [PMID: 38918003 DOI: 10.1667/rade-24-00039.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024]
Abstract
Extracellular vesicles (EVs) have been recognized as a novel way of cell-to-cell communication in the last several decades. It is believed that EVs exert their functions on nearby or distant cells through transfer of the cargo that they carry. In this review, we focus on EVs produced by endothelial cells, with emphasis on their role in hematopoiesis. We first describe how endothelial cells interact with hematopoietic stem/progenitor cells during development and in disease conditions. We then discuss EVs, ranging from their subtypes to isolation methods and analysis of EVs. With the above background information, we next review the literature related to endothelial cell derived EVs (ECEVs), including physiological functions and their clinical uses. In the last sections, we summarize the current results about the effect of ECEVs on hematopoiesis under physiological and stress conditions.
Collapse
Affiliation(s)
| | - Joel R Ross
- Department of Medicine, Duke University, Durham, North Carolina
| | - Nelson J Chao
- Department of Medicine, Duke University, Durham, North Carolina
- Department of Pathology, Duke University, Durham, North Carolina
- Department of Immunology, Duke University, Durham, North Carolina
- Duke Cancer Institute, Duke University, Durham, North Carolina
- Duke Global Health Institute, Duke University, Durham, North Carolina
| | - Benny J Chen
- Department of Medicine, Duke University, Durham, North Carolina
- Department of Immunology, Duke University, Durham, North Carolina
- Duke Fitzpatrick Institute for Photonics, Duke University, Durham, North Carolina
- Duke Regeneration Center, Duke University, Durham, North Carolina
| |
Collapse
|
27
|
Smith C. The potential of zebrafish as drug discovery research tool in immune-mediated inflammatory disease. Inflammopharmacology 2024; 32:2219-2233. [PMID: 38926297 PMCID: PMC11300644 DOI: 10.1007/s10787-024-01511-1] [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: 02/01/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Immune-mediated inflammatory disease (IMID) prevalence is estimated at 3-7% for Westernised populations, with annual incidence reported at almost 1 in 100 people globally. More recently, drug discovery approaches have been evolving towards more targeted therapies with an improved long-term safety profile, while the requirement for individualisation of medicine in complex conditions such as IMIDs, is acknowledged. However, existing preclinical models-such as cellular and in vivo mammalian models-are not ideal for modern drug discovery model requirements, such as real-time in vivo visualisation of drug effects, logistically feasible safety assessment over the course of a lifetime, or dynamic assessment of physiological changes during disease development. Zebrafish share high homology with humans in terms of proteins and disease-causing genes, with high conservation of physiological processes at organ, tissue, cellular and molecular level. These and other unique attributes, such as high fecundity, relative transparency and ease of genetic manipulation, positions zebrafish as the next major role player in IMID drug discovery. This review provides a brief overview of the suitability of this organism as model for human inflammatory disease and summarises the range of approaches used in zebrafish-based drug discovery research. Strengths and limitations of zebrafish as model organism, as well as important considerations in research study design, are discussed. Finally, under-utilised avenues for investigation in the IMID context are highlighted.
Collapse
Affiliation(s)
- Carine Smith
- Experimental Medicine Group, Department of Medicine, Stellenbosch University, Parow, South Africa.
| |
Collapse
|
28
|
Miftari MH, Riste TB, Walther BT. Leukolectins support lifelong innate immunity in lower vertebrates, and reveal dichotomies of several leukocytic lineages. FISH & SHELLFISH IMMUNOLOGY 2024; 151:109685. [PMID: 38857816 DOI: 10.1016/j.fsi.2024.109685] [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: 12/06/2023] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
Innate immunity is vital for animal homeostasis and survival. First-line immuno-defense for fish larvae involves mucus enriched with leukolectin (LL) secreted by dermal lectocytes. Later during the critical transition from yolk-nutrition to feeding, additional larval immuno-protection in zebrafish (zF) is provided by macrophages containing LL (lectophages). This work investigated new LL-expression in embryos and in blood, structures of fish leukocytic LL and LL-genes, and LL-presence in chicken leukocytes. In zF-embryos, lectophages appear ∼10 hpf, while later, cells co-expressing myeloperoxidase- and LL-mRNA were detected (∼19 hpf). Furthermore, protein-extracts of Atlantic salmon (Ssal) leukocytes contained LL-proteins, compartmentalized in the cytosol. Cloning and sequencing revealed 94 % nt-sequence identity between variants of Ssal-leukolectins. Highly conserved LLs allowed production of epitope-specific anti-LL IgGs. Immuno-fluorescence-analysis demonstrated that most Ssal-bloodcells were LL-negative, but both some large cells with protrusions and some small, rounded cells did express LL. Immunoperoxidase-staining method confirmed LL-expression in some Ssal-leukocytes, identified as macrophages, PMN-leukocytes, thrombocytes and dendritic cells. However, closer examination revealed a dichotomy of these cell-categories into either LL-positive, or LL-negative variants. In situ hybridization demonstrated profuse LL-expression in Ssal head kidney interstitial tissue, while LL-transcripts were absent in large kidney tubules. Both hematopoietic (non-pigmented) marrow cells and melano-macrophages expressed LL-mRNA, implying that leukolectins provide lifelong innate immuno-protection. PCR-amplification using Ssal-leukocytic DNA as template, and direct sequencing yielded a leukocytic ll-gene. Some cells in salmon, cod, halibut, oikopleura and zebrafish embryos express LL-proteins and/or LL-mRNA, and LL-mRNA is detected in salmon, cod and chicken leukocytes. However, current genomes for these species lack recognizable LL-loci except the Ssal_v3.1 Genome-assembly. The data demonstrate an unexpected dichotomy of some leukocyte lineages into LL-positive or LL-negative cell-variants. Such dichotomies suggest exploring differential impacts from the duplicated leukocyte-lineages in health and disease.
Collapse
Affiliation(s)
- Mirushe H Miftari
- Department of Molecular Biology, University of Bergen, N-5020, Bergen, Norway
| | - Tonje B Riste
- Department of Molecular Biology, University of Bergen, N-5020, Bergen, Norway
| | - Bernt T Walther
- Department of Molecular Biology, University of Bergen, N-5020, Bergen, Norway.
| |
Collapse
|
29
|
Larionov A, Hammer CM, Fiedler K, Filgueira L. Dynamics of Endothelial Cell Diversity and Plasticity in Health and Disease. Cells 2024; 13:1276. [PMID: 39120307 PMCID: PMC11312403 DOI: 10.3390/cells13151276] [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: 06/27/2024] [Revised: 07/19/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
Endothelial cells (ECs) are vital structural units of the cardiovascular system possessing two principal distinctive properties: heterogeneity and plasticity. Endothelial heterogeneity is defined by differences in tissue-specific endothelial phenotypes and their high predisposition to modification along the length of the vascular bed. This aspect of heterogeneity is closely associated with plasticity, the ability of ECs to adapt to environmental cues through the mobilization of genetic, molecular, and structural alterations. The specific endothelial cytoarchitectonics facilitate a quick structural cell reorganization and, furthermore, easy adaptation to the extrinsic and intrinsic environmental stimuli, known as the epigenetic landscape. ECs, as universally distributed and ubiquitous cells of the human body, play a role that extends far beyond their structural function in the cardiovascular system. They play a crucial role in terms of barrier function, cell-to-cell communication, and a myriad of physiological and pathologic processes. These include development, ontogenesis, disease initiation, and progression, as well as growth, regeneration, and repair. Despite substantial progress in the understanding of endothelial cell biology, the role of ECs in healthy conditions and pathologies remains a fascinating area of exploration. This review aims to summarize knowledge and concepts in endothelial biology. It focuses on the development and functional characteristics of endothelial cells in health and pathological conditions, with a particular emphasis on endothelial phenotypic and functional heterogeneity.
Collapse
Affiliation(s)
- Alexey Larionov
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Christian Manfred Hammer
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| | - Klaus Fiedler
- Independent Researcher, CH-1700 Fribourg, Switzerland;
| | - Luis Filgueira
- Faculty of Science and Medicine, Anatomy, University of Fribourg, Route Albert-Gockel 1, CH-1700 Fribourg, Switzerland; (C.M.H.); (L.F.)
| |
Collapse
|
30
|
Pendse S, Loeffler D. Decoding Clonal Hematopoiesis: Emerging Themes and Novel Mechanistic Insights. Cancers (Basel) 2024; 16:2634. [PMID: 39123361 PMCID: PMC11311828 DOI: 10.3390/cancers16152634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Clonal hematopoiesis (CH), the relative expansion of mutant clones, is derived from hematopoietic stem cells (HSCs) with acquired somatic or cytogenetic alterations that improve cellular fitness. Individuals with CH have a higher risk for hematological and non-hematological diseases, such as cardiovascular disease, and have an overall higher mortality rate. Originally thought to be restricted to a small fraction of elderly people, recent advances in single-cell sequencing and bioinformatics have revealed that CH with multiple expanded mutant clones is universal in the elderly population. Just a few years ago, phylogenetic reconstruction across the human lifespan and novel sensitive sequencing techniques showed that CH can start earlier in life, decades before it was thought possible. These studies also suggest that environmental factors acting through aberrant inflammation might be a common theme promoting clonal expansion and disease progression. However, numerous aspects of this phenomenon remain to be elucidated and the precise mechanisms, context-specific drivers, and pathways of clonal expansion remain to be established. Here, we review our current understanding of the cellular mechanisms driving CH and specifically focus on how pro-inflammatory factors affect normal and mutant HSC fates to promote clonal selection.
Collapse
Affiliation(s)
- Shalmali Pendse
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Pathology & Laboratory Medicine, The University of Tennessee, Memphis, TN 37996, USA
| | - Dirk Loeffler
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Pathology & Laboratory Medicine, The University of Tennessee, Memphis, TN 37996, USA
| |
Collapse
|
31
|
Morganti C, Bonora M, Ito K. Metabolism and HSC fate: what NADPH is made for. Trends Cell Biol 2024:S0962-8924(24)00141-7. [PMID: 39054107 PMCID: PMC11757803 DOI: 10.1016/j.tcb.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial metabolism plays a central role in the regulation of hematopoietic stem cell (HSC) biology. Mitochondrial fatty acid oxidation (FAO) is pivotal in controlling HSC self-renewal and differentiation. Herein, we discuss recent evidence suggesting that NADPH generated in the mitochondria can influence the fate of HSCs. Although NADPH has multiple functions, HSCs show high levels of NADPH that are preferentially used for cholesterol biosynthesis. Endogenous cholesterol supports the biogenesis of extracellular vesicles (EVs), which are essential for maintaining HSC properties. We also highlight the significance of EVs in hematopoiesis through autocrine signaling. Elucidating the mitochondrial NADPH-cholesterol axis as part of the metabolic requirements of healthy HSCs will facilitate the development of new therapies for hematological disorders.
Collapse
Affiliation(s)
- Claudia Morganti
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| | - Massimo Bonora
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| | - Keisuke Ito
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Departments of Oncology and Medicine, Albert Einstein College of Medicine-Montefiore Health System, Bronx, NY 10461, USA.
| |
Collapse
|
32
|
Das A, Smith RJ, Andreadis ST. Harnessing the potential of monocytes/macrophages to regenerate tissue-engineered vascular grafts. Cardiovasc Res 2024; 120:839-854. [PMID: 38742656 PMCID: PMC11218695 DOI: 10.1093/cvr/cvae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 05/16/2024] Open
Abstract
Cell-free tissue-engineered vascular grafts provide a promising alternative to treat cardiovascular disease, but timely endothelialization is essential for ensuring patency and proper functioning post-implantation. Recent studies from our lab showed that blood cells like monocytes (MCs) and macrophages (Mϕ) may contribute directly to cellularization and regeneration of bioengineered arteries in small and large animal models. While MCs and Mϕ are leucocytes that are part of the innate immune response, they share common developmental origins with endothelial cells (ECs) and are known to play crucial roles during vessel formation (angiogenesis) and vessel repair after inflammation/injury. They are highly plastic cells that polarize into pro-inflammatory and anti-inflammatory phenotypes upon exposure to cytokines and differentiate into other cell types, including EC-like cells, in the presence of appropriate chemical and mechanical stimuli. This review focuses on the developmental origins of MCs and ECs; the role of MCs and Mϕ in vessel repair/regeneration during inflammation/injury; and the role of chemical signalling and mechanical forces in Mϕ inflammation that mediates vascular graft regeneration. We postulate that comprehensive understanding of these mechanisms will better inform the development of strategies to coax MCs/Mϕ into endothelializing the lumen and regenerate the smooth muscle layers of cell-free bioengineered arteries and veins that are designed to treat cardiovascular diseases and perhaps the native vasculature as well.
Collapse
Affiliation(s)
- Arundhati Das
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
| | - Randall J Smith
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
| | - Stelios T Andreadis
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, 908 Furnas Hall, Buffalo, NY 14260-4200, USA
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, 332 Bonner Hall, Buffalo, NY 14260-1920, USA
- Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, 701 Ellicott St, Buffalo, NY 14203, USA
- Cell, Gene and Tissue Engineering (CGTE) Center, University at Buffalo, The State University of New York, 813 Furnas Hall, Buffalo, NY 14260-4200, USA
| |
Collapse
|
33
|
Bello AB, Canlas KKV, Kim D, Park H, Lee SH. Stepwise dual-release microparticles of BMP-4 and SCF in induced pluripotent stem cell spheroids enhance differentiation into hematopoietic stem cells. J Control Release 2024; 371:386-405. [PMID: 38844177 DOI: 10.1016/j.jconrel.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.
Collapse
Affiliation(s)
- Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea; School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea
| | | | - Deogil Kim
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06911, Republic of Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul 04620, Republic of Korea.
| |
Collapse
|
34
|
Read NE, Wilson HM. Recent Developments in the Role of Protein Tyrosine Phosphatase 1B (PTP1B) as a Regulator of Immune Cell Signalling in Health and Disease. Int J Mol Sci 2024; 25:7207. [PMID: 39000313 PMCID: PMC11241678 DOI: 10.3390/ijms25137207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a non-receptor tyrosine phosphatase best known for its role in regulating insulin and leptin signalling. Recently, knowledge on the role of PTP1B as a major regulator of multiple signalling pathways involved in cell growth, proliferation, viability and metabolism has expanded, and PTP1B is recognised as a therapeutic target in several human disorders, including diabetes, obesity, cardiovascular diseases and hematopoietic malignancies. The function of PTP1B in the immune system was largely overlooked until it was discovered that PTP1B negatively regulates the Janus kinase-a signal transducer and activator of the transcription (JAK/STAT) signalling pathway, which plays a significant role in modulating immune responses. PTP1B is now known to determine the magnitude of many signalling pathways that drive immune cell activation and function. As such, PTP1B inhibitors are being developed and tested in the context of inflammation and autoimmune diseases. Here, we provide an up-to-date summary of the molecular role of PTP1B in regulating immune cell function and how targeting its expression and/or activity has the potential to change the outcomes of immune-mediated and inflammatory disorders.
Collapse
Affiliation(s)
- Neve E Read
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Heather M Wilson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK
| |
Collapse
|
35
|
Chauhan W, Sudharshan SJ, Kafle S, Zennadi R. SnoRNAs: Exploring Their Implication in Human Diseases. Int J Mol Sci 2024; 25:7202. [PMID: 39000310 PMCID: PMC11240930 DOI: 10.3390/ijms25137202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Small nucleolar RNAs (snoRNAs) are earning increasing attention from research communities due to their critical role in the post-transcriptional modification of various RNAs. These snoRNAs, along with their associated proteins, are crucial in regulating the expression of a vast array of genes in different human diseases. Primarily, snoRNAs facilitate modifications such as 2'-O-methylation, N-4-acetylation, and pseudouridylation, which impact not only ribosomal RNA (rRNA) and their synthesis but also different RNAs. Functionally, snoRNAs bind with core proteins to form small nucleolar ribonucleoproteins (snoRNPs). These snoRNAs then direct the protein complex to specific sites on target RNA molecules where modifications are necessary for either standard cellular operations or the regulation of pathological mechanisms. At these targeted sites, the proteins coupled with snoRNPs perform the modification processes that are vital for controlling cellular functions. The unique characteristics of snoRNAs and their involvement in various non-metabolic and metabolic diseases highlight their potential as therapeutic targets. Moreover, the precise targeting capability of snoRNAs might be harnessed as a molecular tool to therapeutically address various disease conditions. This review delves into the role of snoRNAs in health and disease and explores the broad potential of these snoRNAs as therapeutic agents in human pathologies.
Collapse
Affiliation(s)
| | | | | | - Rahima Zennadi
- Department of Physiology, University of Tennessee Health Science Center, 71 S. Manassas St., Memphis, TN 38103, USA; (W.C.); (S.S.); (S.K.)
| |
Collapse
|
36
|
Feng C, Tie R, Xin S, Chen Y, Li S, Chen Y, Hu X, Zhou Y, Liu Y, Hu Y, Hu Y, Pan H, Wu Z, Chao H, Zhang S, Ni Q, Huang J, Luo W, Huang H, Chen M. Systematic single-cell analysis reveals dynamic control of transposable element activity orchestrating the endothelial-to-hematopoietic transition. BMC Biol 2024; 22:143. [PMID: 38937802 PMCID: PMC11209969 DOI: 10.1186/s12915-024-01939-5] [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: 08/02/2023] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND The endothelial-to-hematopoietic transition (EHT) process during definitive hematopoiesis is highly conserved in vertebrates. Stage-specific expression of transposable elements (TEs) has been detected during zebrafish EHT and may promote hematopoietic stem cell (HSC) formation by activating inflammatory signaling. However, little is known about how TEs contribute to the EHT process in human and mouse. RESULTS We reconstructed the single-cell EHT trajectories of human and mouse and resolved the dynamic expression patterns of TEs during EHT. Most TEs presented a transient co-upregulation pattern along the conserved EHT trajectories, coinciding with the temporal relaxation of epigenetic silencing systems. TE products can be sensed by multiple pattern recognition receptors, triggering inflammatory signaling to facilitate HSC emergence. Interestingly, we observed that hypoxia-related signals were enriched in cells with higher TE expression. Furthermore, we constructed the hematopoietic cis-regulatory network of accessible TEs and identified potential TE-derived enhancers that may boost the expression of specific EHT marker genes. CONCLUSIONS Our study provides a systematic vision of how TEs are dynamically controlled to promote the hematopoietic fate decisions through transcriptional and cis-regulatory networks, and pre-train the immunity of nascent HSCs.
Collapse
Affiliation(s)
- Cong Feng
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310058, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China
- Department of Hematology, The Second Clinical Medical College of Shanxi Medical University, Shanxi Medical University, Taiyuan, 030000, China
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Linhai, 317000, China
| | - Saige Xin
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhao Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Sida Li
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yifan Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaotian Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yincong Zhou
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yongjing Liu
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yueming Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanshi Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hang Pan
- Department of Veterinary Medicine, Zhejiang University College of Animal Sciences, Hangzhou, 310058, China
| | - Zexu Wu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haoyu Chao
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shilong Zhang
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingyang Ni
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jinyan Huang
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Wenda Luo
- Department of Hematology-Oncology, Taizhou Hospital of Zhejiang Province, Linhai, 317000, China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310058, China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, 310058, China.
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- Bioinformatics Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
37
|
Vicente ATS, Salvador JAR. PROteolysis-Targeting Chimeras (PROTACs) in leukemia: overview and future perspectives. MedComm (Beijing) 2024; 5:e575. [PMID: 38845697 PMCID: PMC11154823 DOI: 10.1002/mco2.575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Leukemia is a heterogeneous group of life-threatening malignant disorders of the hematopoietic system. Immunotherapy, radiotherapy, stem cell transplantation, targeted therapy, and chemotherapy are among the approved leukemia treatments. Unfortunately, therapeutic resistance, side effects, relapses, and long-term sequelae occur in a significant proportion of patients and severely compromise the treatment efficacy. The development of novel approaches to improve outcomes is therefore an unmet need. Recently, novel leukemia drug discovery strategies, including targeted protein degradation, have shown potential to advance the field of personalized medicine for leukemia patients. Specifically, PROteolysis-TArgeting Chimeras (PROTACs) are revolutionary compounds that allow the selective degradation of a protein by the ubiquitin-proteasome system. Developed against a wide range of cancer targets, they show promising potential in overcoming many of the drawbacks associated with conventional therapies. Following the exponential growth of antileukemic PROTACs, this article reviews PROTAC-mediated degradation of leukemia-associated targets. Chemical structures, in vitro and in vivo activities, pharmacokinetics, pharmacodynamics, and clinical trials of PROTACs are critically discussed. Furthermore, advantages, challenges, and future perspectives of PROTACs in leukemia are covered, in order to understand the potential that these novel compounds may have as future drugs for leukemia treatment.
Collapse
Affiliation(s)
- André T. S. Vicente
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical ChemistryFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
- Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Center for Innovative Biomedicine and Biotechnology (CIBB)University of CoimbraCoimbraPortugal
| |
Collapse
|
38
|
Brix A, Belleri L, Pezzotta A, Pettinato E, Mazzola M, Zoccolillo M, Marozzi A, Monteiro R, Del Bene F, Mortellaro A, Pistocchi A. ADA2 regulates inflammation and hematopoietic stem cell emergence via the A 2bR pathway in zebrafish. Commun Biol 2024; 7:615. [PMID: 38777862 PMCID: PMC11111730 DOI: 10.1038/s42003-024-06286-3] [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: 09/28/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Deficiency of adenosine deaminase 2 (DADA2) is an inborn error of immunity caused by loss-of-function mutations in the adenosine deaminase 2 (ADA2) gene. Clinical manifestations of DADA2 include vasculopathy and immuno-hematological abnormalities, culminating in bone marrow failure. A major gap exists in our knowledge of the regulatory functions of ADA2 during inflammation and hematopoiesis, mainly due to the absence of an ADA2 orthologue in rodents. Exploring these mechanisms is essential for understanding disease pathology and developing new treatments. Zebrafish possess two ADA2 orthologues, cecr1a and cecr1b, with the latter showing functional conservation with human ADA2. We establish a cecr1b-loss-of-function zebrafish model that recapitulates the immuno-hematological and vascular manifestations observed in humans. Loss of Cecr1b disrupts hematopoietic stem cell specification, resulting in defective hematopoiesis. This defect is caused by induced inflammation in the vascular endothelium. Blocking inflammation, pharmacological modulation of the A2r pathway, or the administration of the recombinant human ADA2 corrects these defects, providing insights into the mechanistic link between ADA2 deficiency, inflammation and immuno-hematological abnormalities. Our findings open up potential therapeutic avenues for DADA2 patients.
Collapse
Affiliation(s)
- Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Laura Belleri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Emanuela Pettinato
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Mara Mazzola
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Matteo Zoccolillo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Anna Marozzi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Rui Monteiro
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, Edgbaston, B15 2TTB, UK
| | - Filippo Del Bene
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alessandra Mortellaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy.
| |
Collapse
|
39
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
40
|
Xu Y, Guo R, Huang T, Guo C. miRNA-7145-cuedc2 axis controls hematopoiesis through JAK1/STAT3 signaling pathway. Cell Death Discov 2024; 10:209. [PMID: 38697957 PMCID: PMC11066045 DOI: 10.1038/s41420-024-01977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Hematopoiesis ensures tissue oxygenation, and remodeling as well as immune protection in vertebrates. During embryogenesis, hemangioblasts are the source of all blood cells. Gata1a and pu.1 are co-expressed in hemangioblasts before hemangioblasts are differentiated into blood cells. However, the genes that determine the differentiation of hemangioblasts into myeloid or erythroid cell lineages have not been fully uncovered. Here we showed that miRNA-7145, a miRNA with previously unknown function, was enriched in erythrocytes at the definitive wave, but not expressed in myeloid cells. Overexpression and loss-of-function analysis of miRNA-7145 revealed that miRNA-7145 functions as a strong inhibitor for myeloid progenitor cell differentiation while driving erythropoiesis during the primitive wave. Furthermore, we confirmed that cuedc2 is one of miRNA-7145 targeted-genes. Overexpression or knock-down of cuedc2 partially rescues the phenotype caused by miRNA-7145 overexpression or loss-of-function. As well, overexpression and loss-of-function analysis of cuedc2 showed that cuedc2 is required for myelopoiesis at the expense of erythropoiesis. Finally, we found that overexpression of zebrafish cuedc2 in 293 T cell inhibits the JAK1/STAT3 signaling pathway. Collectively, our results uncover a previously unknown miRNA-7145-cuedc2 axis, which regulate hematopoiesis through inhibiting the JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yongsheng Xu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| | - Rui Guo
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Tao Huang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Chunming Guo
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
41
|
Speirs ZC, Loynes CA, Mathiessen H, Elks PM, Renshaw SA, Jørgensen LVG. What can we learn about fish neutrophil and macrophage response to immune challenge from studies in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2024; 148:109490. [PMID: 38471626 DOI: 10.1016/j.fsi.2024.109490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
Fish rely, to a high degree, on the innate immune system to protect them against the constant exposure to potential pathogenic invasion from the surrounding water during homeostasis and injury. Zebrafish larvae have emerged as an outstanding model organism for immunity. The cellular component of zebrafish innate immunity is similar to the mammalian innate immune system and has a high degree of sophistication due to the needs of living in an aquatic environment from early embryonic stages of life. Innate immune cells (leukocytes), including neutrophils and macrophages, have major roles in protecting zebrafish against pathogens, as well as being essential for proper wound healing and regeneration. Zebrafish larvae are visually transparent, with unprecedented in vivo microscopy opportunities that, in combination with transgenic immune reporter lines, have permitted visualisation of the functions of these cells when zebrafish are exposed to bacterial, viral and parasitic infections, as well as during injury and healing. Recent findings indicate that leukocytes are even more complex than previously anticipated and are essential for inflammation, infection control, and subsequent wound healing and regeneration.
Collapse
Affiliation(s)
- Zoë C Speirs
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Catherine A Loynes
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Heidi Mathiessen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark
| | - Philip M Elks
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Stephen A Renshaw
- The Bateson Centre, School of Medicine and Population Health, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Louise von Gersdorff Jørgensen
- Laboratory of Experimental Fish Models, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg C., Denmark.
| |
Collapse
|
42
|
Guo C, Lv X, Zhang Q, Yi L, Ren Y, Li Z, Yan J, Zheng S, Sun M, Liu S. CRKL but not CRKII contributes to hemin-induced erythroid differentiation of CML. J Cell Mol Med 2024; 28:e18308. [PMID: 38683131 PMCID: PMC11057422 DOI: 10.1111/jcmm.18308] [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: 07/06/2023] [Revised: 11/22/2023] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Destruction of erythropoiesis process leads to various diseases, including thrombocytopenia, anaemia, and leukaemia. miR-429-CT10 regulation of kinase-like (CRKL) axis involved in development, progression and metastasis of cancers. However, the exact role of miR-429-CRKL axis in leukaemic cell differentiation are still unknown. The current work aimed to uncover the effect of miR-429-CRKL axis on erythropoiesis. In the present study, CRKL upregulation was negatively correlated with miR-429 downregulation in both chronic myeloid leukaemia (CML) patient and CR patient samples. Moreover, CRKL expression level was significantly decreased while miR-429 expression level was increased during the erythroid differentiation of K562 cells following hemin treatment. Functional investigations revealed that overexpression and knockdown of CRKL was remarkably effective in suppressing and promoting hemin-induced erythroid differentiation of K562 cells, whereas, miR-429 exhibited opposite effects to CRKL. Mechanistically, miR-429 regulates erythroid differentiation of K562 cells by downregulating CRKL via selectively targeting CRKL-3'-untranslated region (UTR) through Raf/MEK/ERK pathway. Conversely, CRKII had no effect on erythroid differentiation of K562 cells. Taken together, our data demonstrated that CRKL (but not CRKII) and miR-429 contribute to development, progression and erythropoiesis of CML, miR-429-CRKL axis regulates erythropoiesis of K562 cells via Raf/MEK/ERK pathway, providing novel insights into effective diagnosis and therapy for CML patients.
Collapse
MESH Headings
- Humans
- 3' Untranslated Regions
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Cell Differentiation/drug effects
- Erythroid Cells/metabolism
- Erythroid Cells/drug effects
- Erythroid Cells/pathology
- Erythroid Cells/cytology
- Erythropoiesis/genetics
- Erythropoiesis/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Hemin/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- MAP Kinase Signaling System/drug effects
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Proto-Oncogene Proteins c-crk/metabolism
- Proto-Oncogene Proteins c-crk/genetics
Collapse
Affiliation(s)
- Chunmei Guo
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Xinxin Lv
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Qiuling Zhang
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Lina Yi
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Yingying Ren
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Zhaopeng Li
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Jinsong Yan
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical UniversityInstitute of Stem Cell Transplantation of Dalian Medical UniversityDalianLiaoningChina
| | - Shanliang Zheng
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Ming‐Zhong Sun
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Shuqing Liu
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
43
|
Pomreinke AP, Müller P. Zebrafish nampt-a mutants are viable despite perturbed primitive hematopoiesis. Hereditas 2024; 161:14. [PMID: 38685093 PMCID: PMC11057069 DOI: 10.1186/s41065-024-00318-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 04/17/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Nicotinamide phosphoribosyltransferase (Nampt) is required for recycling NAD+ in numerous cellular contexts. Morpholino-based knockdown of zebrafish nampt-a has been shown to cause abnormal development and defective hematopoiesis concomitant with decreased NAD+ levels. However, surprisingly, nampt-a mutant zebrafish were recently found to be viable, suggesting a discrepancy between the phenotypes in knockdown and knockout conditions. Here, we address this discrepancy by directly comparing loss-of-function approaches that result in identical defective transcripts in morphants and mutants. RESULTS Using CRISPR/Cas9-mediated mutagenesis, we generated nampt-a mutant lines that carry the same mis-spliced mRNA as nampt-a morphants. Despite reduced NAD+ levels and perturbed expression of specific blood markers, nampt-a mutants did not display obvious developmental defects and were found to be viable. In contrast, injection of nampt-a morpholinos into wild-type or mutant nampt-a embryos caused aberrant phenotypes. Moreover, nampt-a morpholinos caused additional reduction of blood-related markers in nampt-a mutants, suggesting that the defects observed in nampt-a morphants can be partially attributed to off-target effects of the morpholinos. CONCLUSIONS Our findings show that zebrafish nampt-a mutants are viable despite reduced NAD+ levels and a perturbed hematopoietic gene expression program, indicating strong robustness of primitive hematopoiesis during early embryogenesis.
Collapse
Affiliation(s)
- Autumn Penecilla Pomreinke
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
- University of Hohenheim, Stuttgart, Germany
| | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany.
- University of Konstanz, Konstanz, Germany.
| |
Collapse
|
44
|
Xiao S, Chen H, Bai Y, Zhang ZY, Liu Y. Targeting PRL phosphatases in hematological malignancies. Expert Opin Ther Targets 2024; 28:259-271. [PMID: 38653737 PMCID: PMC12050007 DOI: 10.1080/14728222.2024.2344695] [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: 10/13/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Phosphatase of regenerating liver (PRL) family proteins, also known as protein tyrosine phosphatase 4A (PTP4A), have been implicated in many types of cancers. The PRL family of phosphatases consists of three members, PRL1, PRL2, and PRL3. PRLs have been shown to harbor oncogenic potentials and are highly expressed in a variety of cancers. Given their roles in cancer progression and metastasis, PRLs are potential targets for anticancer therapies. However, additional studies are needed to be performed to fully understand the roles of PRLs in blood cancers. AREAS COVERED In this review, we will summarize recent studies of PRLs in normal and malignant hematopoiesis, the role of PRLs in regulating various signaling pathways, and the therapeutic potentials of targeting PRLs in hematological malignancies. We will also discuss how to improve current PRL inhibitors for cancer treatment. EXPERT OPINION Although PRL inhibitors show promising therapeutic effects in preclinical studies of different types of cancers, moving PRL inhibitors from bench to bedside is still challenging. More potent and selective PRL inhibitors are needed to target PRLs in hematological malignancies and improve treatment outcomes.
Collapse
Affiliation(s)
- Shiyu Xiao
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| | - Hongxia Chen
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Hematology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Yunpeng Bai
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Institute for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, USA
| | - Yan Liu
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, USA
| |
Collapse
|
45
|
Welsh AM, Muljo SA. Post-transcriptional (re)programming of B lymphocyte development: From bench to bedside? Adv Immunol 2024; 161:85-108. [PMID: 38763703 DOI: 10.1016/bs.ai.2024.03.003] [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: 05/21/2024]
Abstract
Hematopoiesis, a process which generates blood and immune cells, changes significantly during mammalian development. Definitive hematopoiesis is marked by the emergence of long-term hematopoietic stem cells (HSCs). Here, we will focus on the post-transcriptional differences between fetal liver (FL) and adult bone marrow (ABM) HSCs. It remains unclear how or why exactly FL HSCs transition to ABM HSCs, but we aim to leverage their differences to revive an old idea: in utero HSC transplantation. Unexpectedly, the expression of certain RNA-binding proteins (RBPs) play an important role in HSC specification, and can be employed to convert or reprogram adult HSCs back to a fetal-like state. Among other features, FL HSCs have a broad differentiation capacity that includes the ability to regenerate both conventional B and T cells, as well as innate-like or unconventional lymphocytes such as B-1a and marginal zone B (MzB) cells. This chapter will focus on RNA binding proteins, namely LIN28B and IGF2BP3, that are expressed during fetal life and how they promote B-1a cell development. Furthermore, this chapter considers a potential clinical application of synthetic co-expression of LIN28B and IGF2BP3 in HSCs.
Collapse
Affiliation(s)
- Alia M Welsh
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stefan A Muljo
- Integrative Immunobiology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.
| |
Collapse
|
46
|
Zhang A, Lu J, Feng S, Yu H, Yu T, Zhao S, Chen K, Huang Z, Xu J, Qu JY, Wen Z. Fli1 acts in parallel with Pu.1 to control macrophage and neutrophil fate in zebrafish. J Genet Genomics 2024; 51:359-362. [PMID: 37944856 DOI: 10.1016/j.jgg.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 11/03/2023] [Accepted: 11/04/2023] [Indexed: 11/12/2023]
Affiliation(s)
- Ao Zhang
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jingao Lu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Shachuan Feng
- Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Huapeng Yu
- Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Tao Yu
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Shizheng Zhao
- Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong 518036, China
| | - Kemin Chen
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Zhibin Huang
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Jianan Y Qu
- Department of Electronic and Computer Engineering, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Zilong Wen
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Department of Immunology and Microbiology, School of Life Science, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518067, China.
| |
Collapse
|
47
|
Taj T, Chen J, Rodopoulou S, Strak M, de Hoogh K, Poulsen AH, Andersen ZJ, Bellander T, Brandt J, Zitt E, Fecht D, Forastiere F, Gulliver J, Hertel O, Hoffmann B, Hvidtfeldt UA, Jørgensen JT, Katsouyanni K, Ketzel M, Lager A, Leander K, Liu S, Ljungman P, Severi G, Besson C, Magnusson PKE, Nagel G, Pershagen G, Peters A, Rizzuto D, Samoli E, Sørensen M, Stafoggia M, Tjønneland A, Weinmayr G, Wolf K, Brunekreef B, Hoek G, Raaschou-Nielsen O. Long-term exposure to ambient air pollution and risk of leukemia and lymphoma in a pooled European cohort. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 343:123097. [PMID: 38065336 DOI: 10.1016/j.envpol.2023.123097] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/08/2023] [Accepted: 12/02/2023] [Indexed: 12/19/2023]
Abstract
Leukemia and lymphoma are the two most common forms of hematologic malignancy, and their etiology is largely unknown. Pathophysiological mechanisms suggest a possible association with air pollution, but little empirical evidence is available. We aimed to investigate the association between long-term residential exposure to outdoor air pollution and risk of leukemia and lymphoma. We pooled data from four cohorts from three European countries as part of the "Effects of Low-level Air Pollution: a Study in Europe" (ELAPSE) collaboration. We used Europe-wide land use regression models to assess annual mean concentrations of fine particulate matter (PM2.5), nitrogen dioxide (NO2), black carbon (BC) and ozone (O3) at residences. We also estimated concentrations of PM2.5 elemental components: copper (Cu), iron (Fe), zinc (Zn); sulfur (S); nickel (Ni), vanadium (V), silicon (Si) and potassium (K). We applied Cox proportional hazards models to investigate the associations. Among the study population of 247,436 individuals, 760 leukemia and 1122 lymphoma cases were diagnosed during 4,656,140 person-years of follow-up. The results showed a leukemia hazard ratio (HR) of 1.13 (95% confidence intervals [CI]: 1.01-1.26) per 10 μg/m3 NO2, which was robust in two-pollutant models and consistent across the four cohorts and according to smoking status. Sex-specific analyses suggested that this association was confined to the male population. Further, the results showed increased lymphoma HRs for PM2.5 (HR = 1.16; 95% CI: 1.02-1.34) and potassium content of PM2.5, which were consistent in two-pollutant models and according to sex. Our results suggest that air pollution at the residence may be associated with adult leukemia and lymphoma.
Collapse
Affiliation(s)
- Tahir Taj
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark.
| | - Jie Chen
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Sophia Rodopoulou
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Maciej Strak
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands; National Institute for Public Health and the Environment, Bilthoven, the Netherlands.
| | - Kees de Hoogh
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland.
| | | | - Zorana J Andersen
- Section of Environment and Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Tom Bellander
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Occupational and Environmental Medicine, Region Stockholm, Stockholm, Sweden.
| | - Jørgen Brandt
- Department of Environmental Science, Aarhus University, Frederiksborgvej 399, DK-4000 Roskilde, Denmark.
| | - Emanuel Zitt
- Agency for Preventive and Social Medicine (aks), Bregenz, Austria; Department of Internal Medicine 3, LKH Feldkirch, Feldkirch, Austria.
| | - Daniela Fecht
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom.
| | - Francesco Forastiere
- Department of Epidemiology, Lazio Region Health Service, ASL Roma 1, Rome, Italy; Environmental Research Group, School of Public Health, Faculty of Medicine, Imperial College, London, United Kingdom.
| | - John Gulliver
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom; Centre for Environmental Health and Sustainability & School of Geography, Geology and the Environment, University of Leicester, Leicester, United Kingdom.
| | - Ole Hertel
- Department of Ecoscience, Aarhus University, Roskilde, Denmark.
| | - Barbara Hoffmann
- Institute for Occupational, Social and Environmental Medicine, Centre for Health and Society, Medical Faculty, Heinrich Heine University Düsseldorf, Germany.
| | | | - Jeanette T Jørgensen
- Section of Environment and Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Klea Katsouyanni
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece; MRC Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom.
| | - Matthias Ketzel
- Department of Environmental Science, Aarhus University, Frederiksborgvej 399, DK-4000 Roskilde, Denmark; Global Centre for Clean Air Research (GCARE), University of Surrey, Guildford GU2 7XH, United Kingdom.
| | - Anton Lager
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.
| | - Karin Leander
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Shuo Liu
- Section of Environment and Health, Department of Public Health, University of Copenhagen, Copenhagen, Denmark.
| | - Petter Ljungman
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Cardiology, Danderyd University Hospital, Stockholm, Sweden.
| | - Gianluca Severi
- University Paris-Saclay, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" Team, CESP UMR1018, 94805, Villejuif, France; Department of Statistics, Computer Science, Applications "G. Parenti" (DISIA), University of Florence, Italy.
| | - Caroline Besson
- University Paris-Saclay, UVSQ, Inserm, Gustave Roussy, "Exposome and Heredity" Team, CESP UMR1018, 94805, Villejuif, France.
| | - Patrik K E Magnusson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Gabriele Nagel
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany; Chair of Epidemiology, Ludwig Maximilians Universität München, Munich, Germany.
| | - Debora Rizzuto
- Department of Neurobiology, Care Sciences, and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden; Stockholm Gerontology Research Center, Stockholm, Sweden.
| | - Evangelia Samoli
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| | - Mette Sørensen
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Roskilde, Denmark.
| | - Massimo Stafoggia
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Epidemiology, Lazio Region Health Service, ASL Roma 1, Rome, Italy.
| | - Anne Tjønneland
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark.
| | - Gudrun Weinmayr
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany.
| | - Kathrin Wolf
- Institute of Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Bert Brunekreef
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Gerard Hoek
- Institute for Risk Assessment Sciences, Utrecht University, Utrecht, the Netherlands.
| | - Ole Raaschou-Nielsen
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark; Department of Environmental Science, Aarhus University, Frederiksborgvej 399, DK-4000 Roskilde, Denmark.
| |
Collapse
|
48
|
Mastrogiovanni M, Martínez-Navarro FJ, Bowman TV, Cayuela ML. Inflammation in Development and Aging: Insights from the Zebrafish Model. Int J Mol Sci 2024; 25:2145. [PMID: 38396822 PMCID: PMC10889087 DOI: 10.3390/ijms25042145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/30/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Zebrafish are an emergent animal model to study human diseases due to their significant genetic similarity to humans, swift development, and genetic manipulability. Their utility extends to the exploration of the involvement of inflammation in host defense, immune responses, and tissue regeneration. Additionally, the zebrafish model system facilitates prompt screening of chemical compounds that affect inflammation. This study explored the diverse roles of inflammatory pathways in zebrafish development and aging. Serving as a crucial model, zebrafish provides insights into the intricate interplay of inflammation in both developmental and aging contexts. The evidence presented suggests that the same inflammatory signaling pathways often play instructive or beneficial roles during embryogenesis and are associated with malignancies in adults.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Francisco Juan Martínez-Navarro
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
| | - Teresa V. Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - María L. Cayuela
- Grupo de Telomerasa, Cáncer y Envejecimiento, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria-Arrixaca, 30120 Murcia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 30100 Murcia, Spain
| |
Collapse
|
49
|
Li G, Sun Y, Kwok I, Yang L, Wen W, Huang P, Wu M, Li J, Huang Z, Liu Z, He S, Peng W, Bei JX, Ginhoux F, Ng LG, Zhang Y. Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish. Nat Commun 2024; 15:811. [PMID: 38280871 PMCID: PMC10821951 DOI: 10.1038/s41467-024-45029-0] [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: 03/06/2023] [Accepted: 01/11/2024] [Indexed: 01/29/2024] Open
Abstract
Eosinophils are a group of granulocytes well known for their capacity to protect the host from parasites and regulate immune function. Diverse biological roles for eosinophils have been increasingly identified, but the developmental pattern and regulation of the eosinophil lineage remain largely unknown. Herein, we utilize the zebrafish model to analyze eosinophilic cell differentiation, distribution, and regulation. By identifying eslec as an eosinophil lineage-specific marker, we establish a Tg(eslec:eGFP) reporter line, which specifically labeled cells of the eosinophil lineage from early life through adulthood. Spatial-temporal analysis of eslec+ cells demonstrates their organ distribution from larval stage to adulthood. By single-cell RNA-Seq analysis, we decipher the eosinophil lineage cells from lineage-committed progenitors to mature eosinophils. Through further genetic analysis, we demonstrate the role of Cebp1 in balancing neutrophil and eosinophil lineages, and a Cebp1-Cebpβ transcriptional axis that regulates the commitment and differentiation of the eosinophil lineage. Cross-species functional comparisons reveals that zebrafish Cebp1 is the functional orthologue of human C/EBPεP27 in suppressing eosinophilopoiesis. Our study characterizes eosinophil development in multiple dimensions including spatial-temporal patterns, expression profiles, and genetic regulators, providing for a better understanding of eosinophilopoiesis.
Collapse
Affiliation(s)
- Gaofei Li
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Yicong Sun
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore
| | - Liting Yang
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Wanying Wen
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Peixian Huang
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Mei Wu
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Jing Li
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Zhibin Huang
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Shuai He
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Wan Peng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Jin-Xin Bei
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, P.R. China
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648, Singapore.
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, P.R. China.
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117543, Singapore.
| | - Yiyue Zhang
- Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China.
- Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, P.R. China.
| |
Collapse
|
50
|
Balaji S, Anbarasu S, Ramaiah S, Anbarasu A. Targeting IKZF1 via HDAC1: Combating Acute Myeloid Leukemia. Integr Biol (Camb) 2024; 16:zyae022. [PMID: 39679961 DOI: 10.1093/intbio/zyae022] [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: 05/21/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Acute myeloid leukemia (AML) accounts for 1.3% of all cancers, with a limited survival of only 30%, and treating AML is a continuous challenge in medicine. IKZF1 is a DNA-binding protein that is highly mutated and undruggable but significant in causing AML. The current study aims to target its transcription factors (TFs) modulating IKZF1 activity. The TF network was constructed and analyzed which revealed a dense Markov cluster (MCL) cluster and five hub genes namely, HDAC1, EP300, CREBBP, TP53, and MYC; the first node clusters were generated for the hub genes. Functional enrichment analysis found AML pathway enriched in all the clusters. Gene ontology terms were majorly related to transcription regulation terms including RNA polymerase transcription regulation, DNA binding activity, DNA templated transcription, and transcription factor binding. Further, the mutation profile of all the TFs found HDAC1 with a very low mutation profile of 0.1% and the survival plot found HDAC1 with a hazard ratio of 1.17 with increased survival upon low expression. Also, among the hub genes, HDAC1 was the only first node interactor with IKZF1. Thus, HDAC1 could be a potential biomarker candidate as well as a key target in treating AML. Insight Box The study has an integrated approach for identifying a potential target through network analysis, functional enrichment analysis, mutation profiling survival prognosis, and target screening. The study employs a better strategy for targeting IKZF1, a significantly upregulated gene in AML by regulating its transcription factors. The analysis revealed a network of TFs regulating IKZF1, among which HDAC1 emerged as a promising candidate due to its low mutation rate, association with better survival outcomes, and direct interaction with IKZF1. This suggests HDAC1 could be a valuable biomarker and therapeutic target for AML treatment.
Collapse
Affiliation(s)
- Sathyanarayan Balaji
- Department of Biotechnology, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
| | - Suvitha Anbarasu
- Department of Biotechnology, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
- Medical and Biological Computing Laboratory, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
| | - Sudha Ramaiah
- Medical and Biological Computing Laboratory, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
- Department of Biosciences, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
| | - Anand Anbarasu
- Department of Biotechnology, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
- Medical and Biological Computing Laboratory, School of Bioscience and Technology (SBST), Vellore Institute of Technology (VIT), Vellore District, Tamil Nadu State, 632014, India
| |
Collapse
|