1
|
Emili E, Pérez-Posada A, Vanni V, Salamanca-Díaz D, Ródriguez-Fernández D, Christodoulou MD, Solana J. Allometry of cell types in planarians by single-cell transcriptomics. SCIENCE ADVANCES 2025; 11:eadm7042. [PMID: 40333969 PMCID: PMC12057665 DOI: 10.1126/sciadv.adm7042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/02/2025] [Indexed: 05/09/2025]
Abstract
Allometry explores the relationship between an organism's body size and its various components, offering insights into ecology, physiology, metabolism, and disease. The cell is the basic unit of biological systems, and yet the study of cell-type allometry remains relatively unexplored. Single-cell RNA sequencing (scRNA-seq) provides a promising tool for investigating cell-type allometry. Planarians, capable of growing and degrowing following allometric scaling rules, serve as an excellent model for these studies. We used scRNA-seq to examine cell-type allometry in asexual planarians of different sizes, revealing that they consist of the same basic cell types but in varying proportions. Notably, the gut basal cells are the most responsive to changes in size, suggesting a role in energy storage. We capture the regulated gene modules of distinct cell types in response to body size. This research sheds light on the molecular and cellular aspects of cell-type allometry in planarians and underscores the utility of scRNA-seq in these investigations.
Collapse
Affiliation(s)
- Elena Emili
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Alberto Pérez-Posada
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
- Department of Biosciences, University of Exeter, Exeter, UK
| | - Virginia Vanni
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
- Department of Biosciences, University of Exeter, Exeter, UK
| | - David Salamanca-Díaz
- Living Systems Institute, University of Exeter, Exeter, UK
- Department of Biosciences, University of Exeter, Exeter, UK
| | | | | | - Jordi Solana
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
- Department of Biosciences, University of Exeter, Exeter, UK
| |
Collapse
|
2
|
Liu Y, Li C, Shen LC, Yan H, Wei G, Gasser RB, Hu X, Song J, Yu DJ. scRCA: A Siamese network-based pipeline for annotating cell types using noisy single-cell RNA-seq reference data. Comput Biol Med 2025; 190:110068. [PMID: 40158457 DOI: 10.1016/j.compbiomed.2025.110068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
Accurate cell type annotation is fundamentally critical for single-cell sequencing (scRNA-seq) data analysis to provide insightful knowledge of tissue-specific cell heterogeneity and cell state transition tracking. Cell type annotation is usually conducted by comparative analysis with known data (i.e., reference) - which contains a presumably accurate representation of cell types. However, this assumption is often problematic, as factors such as human errors in wet-lab experiments and methodological limitations can introduce annotation errors in the reference dataset. As current pipelines for single-cell transcriptomic analysis do not adequately consider this challenge, there is a major demand for constructing a computational pipeline that achieves high-quality cell type annotation using reference datasets containing inherent errors (referred to as "noise" in this study). Here, we built a Siamese network-based pipeline, termed scRCA, to accurately annotate cell types based on noisy reference data. To help users evaluate the reliability of scRCA annotations, an interpreter was also developed to explore the factors underlying the model's predictions. Our experiments demonstrate that, across 14 datasets, scRCA outperformed other widely adopted reference-based methods for cell type annotation. Using an independent dataset of four multiple myeloma patients, we further illustrated that scRCA can distinguish cancerous cells based on gene expression levels and identify genes closely associated with multiple myeloma through scRCA's interpretable module, providing significant information for subsequent clinical treatments. With these advancements, we anticipate that scRCA will serve as a practical reference-based approach for accurate annotating cell type annotation.
Collapse
Affiliation(s)
- Yan Liu
- Department of Computer Science, Yangzhou University, Yangzhou, 225100, China
| | - Chen Li
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia
| | - Long-Chen Shen
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - He Yan
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China
| | - Guo Wei
- School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Robin B Gasser
- Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia
| | - Xiaohua Hu
- Information Department, The First Affiliated Hospital of Naval Military Medical University, Changhai Road 168, Shanghai, 200433, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, 3800, Australia; Monash Data Futures Institute, Monash University, Melbourne, Victoria, 3800, Australia.
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei, Nanjing, 210094, China.
| |
Collapse
|
3
|
Dai X, Li X, Tyshkovskiy A, Zuckerman C, Cheng N, Lin P, Paris D, Qureshi S, Kruglyak L, Mao X, Nandakumar J, Gladyshev VN, Pletcher S, Sobota J, Guo L. Regeneration leads to global tissue rejuvenation in aging sexual planarians. NATURE AGING 2025; 5:780-798. [PMID: 40181188 DOI: 10.1038/s43587-025-00847-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 03/03/2025] [Indexed: 04/05/2025]
Abstract
The possibility of reversing the adverse impacts of aging could significantly reduce age-related diseases and improve quality of life in older populations. Here we report that the sexual lineage of the planarian Schmidtea mediterranea exhibits physiological decline within 18 months of birth, including altered tissue architecture, impaired fertility and motility, and increased oxidative stress. Single-cell profiling of young and older planarian heads uncovered loss of neurons and muscle, increase of glia, and revealed minimal changes in somatic pluripotent stem cells, along with molecular signatures of aging across tissues. Remarkably, amputation followed by regeneration of lost tissues in older planarians led to reversal of these age-associated changes in tissues both proximal and distal to the injury at physiological, cellular and molecular levels. Our work suggests mechanisms of rejuvenation in both new and old tissues concurring with planarian regeneration, which may provide valuable insights for antiaging interventions.
Collapse
Affiliation(s)
- Xiaoting Dai
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Xinghua Li
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cassandra Zuckerman
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Nan Cheng
- Department of Physics, University of Michigan, Ann Arbor, MI, USA
| | - Peter Lin
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - David Paris
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Saad Qureshi
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Leonid Kruglyak
- Department of Human Genetics, Department of Biological Chemistry, Howard Hughes Medical Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiaoming Mao
- Department of Physics, University of Michigan, Ann Arbor, MI, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Scott Pletcher
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Jacob Sobota
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Longhua Guo
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
4
|
Ouyang J, Han G, Chen J, Hu J, Luo L, Zhang H, Lan C, Lu Q, Gou Y, Gu H, Hu Y, Zhang P, Xu A, Huang S. Identification and characterization of a novel ApeC-containing transmembrane protein family in parasitic flatworms. Int J Biol Macromol 2025; 309:142866. [PMID: 40210028 DOI: 10.1016/j.ijbiomac.2025.142866] [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: 11/24/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
The Apextrin C-terminal (ApeC) domain is widely distributed across various animal clades. Several ApeC domains have been identified as carbohydrate-binding domains, contributing to extracellular adhesion and mucosal immunity. However, the functions and evolutionary history of most ApeC-containing protein (ACP) families remain largely unexplored. In this study, we identified 213 ACPs in flatworms (Platyhelminthes), with each species containing one to two such proteins. All flatworm ACPs belong to a unique transmembrane protein family, characterized by a length of 1700-2200 amino acids and a distinctive domain architecture (SP-[Kringle]1-2-[ApeC]7-[EGF]1-4-TM) unlike any found in other phyla. This ACP is conserved across all major parasitic flatworm lineages, including flukes (Trematoda), tapeworms (Cestoda) and monogeneans (Monogenea), despite their diverse morphologies and habitats. Notably, it is also present in one group of free-living flatworms, the planarians (Tricladida), suggesting that this ACP originated in the free-living ancestor of parasitic flatworms. This ACP contains seven consecutive ApeC domains, an unparalleled number among known animal proteins. Five of these ApeC domains are highly divergent, necessitating the definition of a new domain model (ApeC_Pla; Pfam: PF24148) for accurate classification. Structural predictions indicate that these ApeC domains adopt a conserved three β-sheet structure. Furthermore, transcriptomic analysis revealed that flatworm ACPs, along with several important serpins and proteases, are predominantly expressed in parenchymal cells and feeding organs, suggesting that ACPs serve as novel marker genes for parenchymal tissue and may be involved in cell adhesion, oral immunity, and parasite-host interactions. Taken together, our findings indicate that this flatworm ACP represents a promising target for vaccine development and provides key insights into the physiology of the parenchyma, a unique flatworm tissue that functions as a substitute for a body cavity.
Collapse
Affiliation(s)
- Jihua Ouyang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Guangkun Han
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Jinsong Chen
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Jiaxuan Hu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Lei Luo
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Hao Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Chunliu Lan
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Qianyu Lu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Yin Gou
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Hanjie Gu
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Yonghua Hu
- Institute of Tropical Bioscience and Biotechnology, Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya 572024, China
| | - Peng Zhang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Anlong Xu
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China
| | - Shengfeng Huang
- State Key Laboratory of Biocontrol, Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Guangdong Key Laboratory of Pharmaceutical Functional Genes, School of Life Sciences, Sun Yat-sen University, Guangdong, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
5
|
Ling X, Chen G, Liu N, Xu W, Ding M. Integration of Single-Cell RNA Sequencing Data and Bulk Sequencing Data to Characterise the CD8+ T-Cell Exhaustion Mediated Immune Microenvironment in CRC. J Cell Mol Med 2025; 29:e70556. [PMID: 40356050 PMCID: PMC12069026 DOI: 10.1111/jcmm.70556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
CD8+ T cells are crucial for the anti-tumour immune response, and their exhaustion contributes to poor prognosis and limited immunotherapy efficacy in colorectal cancer (CRC). In this study, we examined the immune microenvironment of CRC by integrating single-cell RNA sequencing (scRNA-seq) and bulk sequencing data. T-cell subtypes in tumour tissues were analysed using CellMarker 2.0 and scType, and an intercellular communication network was constructed through CellChat. Our analysis revealed that exhausted CD8+ T cells exhibit strong interactions with epithelial cells, primarily via the MIF-(CD74 + CXCR4), MIF-(CD74 + CD44) and CD99-CD99 pathways. Based on CD8+ T-cell exhaustion markers, we developed a prognostic model using XGBoost, which demonstrated promising predictive capabilities for CRC prognosis and immunotherapy response. Functional assays showed that MIF knock-down significantly inhibited CRC cell proliferation and invasion. Our findings suggest that MIF and CD99 are key regulators of CD8+ T-cell exhaustion in CRC. This study provides novel insights into the mechanisms underlying T-cell exhaustion in CRC and offers potential biomarkers for improving immunotherapy outcomes.
Collapse
Affiliation(s)
- Xiao‐Hua Ling
- Department of GastroenterologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinPeople's Republic of China
| | - Gang Chen
- Department of General SurgeryThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Nan‐Nan Liu
- Department of GastroenterologyThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinPeople's Republic of China
| | - Wen‐Xin Xu
- Department of Intensive Care UnitThe Fourth Affiliated Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| | - Ming‐Feng Ding
- Department of General SurgeryThe Fourth Hospital of Harbin Medical UniversityHarbinHeilongjiangPeople's Republic of China
| |
Collapse
|
6
|
Ma K, Guo F, Li R, Song G, Zhang H, Lu Q, Ma K, Gong S. Knockdown of Atg1 Impairs Brain Regeneration and Downregulates ECM-Related Genes in the Planarian Dugesia japonica. Mol Neurobiol 2025:10.1007/s12035-025-04978-3. [PMID: 40281299 DOI: 10.1007/s12035-025-04978-3] [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: 12/15/2024] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Planarian regeneration is a complex process that involves the precise orchestration of cell proliferation, differentiation, migration, and autophagy. However, the role of autophagy in planarian regeneration remains poorly understood. In this study, we identified autophagy-related gene 1 from the planarian Dugesia japonica (designated as DjAtg1) and investigated its role in planarian brain regeneration. DjAtg1 transcripts are highly expressed in the cephalic ganglia of intact planarians. Following amputation, DjAtg1 is prominently expressed in the newly regenerated brain tissues. Knockdown of DjAtg1 via RNA interference (RNAi) induces head regression, with all RNAi-treated animals regenerating a small triangular-shaped head. Neoblast-marker labeling experiments demonstrate that DjAtg1 knockdown does not affect cell proliferation but impairs neoblast behavior. Notably, RNA-seq reveals that most of these down-regulated transcripts are linked to the extracellular matrix (ECM). Based on our findings and prior literature, we propose that the DjAtg1-mediated secretory pathway is essential for ECM remodeling. DjAtg1 knockdown disrupts the secretory pathway, which feedback-inhibits the expression of ECM-related genes. Our work provides new insights into the non-canonical role of autophagy in regulating of ECM remodeling during planarian regeneration.
Collapse
Affiliation(s)
- Kexue Ma
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China.
- Henan Province Engineering Research Center of Nutrition and Health, Luohe, 462002, China.
| | - Fangying Guo
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Rui Li
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Gege Song
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hecai Zhang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Qiong Lu
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China
| | - Keshi Ma
- College of Life Sciences and Agronomy, Zhoukou Normal University, Zhoukou, 466001, China
| | - Shaoqing Gong
- Department of Basic Medicine, Luohe Medical College, Luohe, 462002, China.
| |
Collapse
|
7
|
Wang KT, Chen YC, Tsai FY, Judy CP, Adler CE. Pluripotent Stem Cell Plasticity is Sculpted by a Slit-Independent Robo Pathway in a Regenerative Animal. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.14.648795. [PMID: 40376085 PMCID: PMC12080947 DOI: 10.1101/2025.04.14.648795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Whole-body regeneration requires adult stem cells with high plasticity to differentiate into missing cell types. Planarians possess a unique configuration of organs embedded in a vast pool of pluripotent stem cells. How stem cells integrate positional information with discrete fates remains unknown. Here, we use the planarian pharynx to define the cell fates that depend on the pioneer transcription factor FoxA. We find that Roundabout receptor RoboA suppresses aberrant pharynx cell fates by altering foxA expression, independent of the canonical ligand Slit. An RNAi screen for extracellular proteins identifies Anosmin-1 as a potential partner of RoboA. Perturbing global patterning demonstrates that roboA / anosmin-1 functions locally in the brain. By contrast, altering pharynx fate with foxA knockdown induces head-specific neurons in the pharynx, indicating a latent plasticity of stem cells. Our data links critical extracellular cues with cell fate decisions of highly plastic stem cells, ensuring the fidelity of organ regeneration.
Collapse
|
8
|
Chai C, Sultan E, Sarkar SR, Zhong L, Sarfati DN, Gershoni-Yahalom O, Jacobs-Wagner C, Rosental B, Wang B. Explosive cytotoxicity of 'ruptoblasts' bridges hormonal surveillance and immune defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645876. [PMID: 40236000 PMCID: PMC11996342 DOI: 10.1101/2025.03.28.645876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytotoxic killing is an essential immune function, yet its cellular mechanisms have been characterized in only a few model species. Here, we show that planarian flatworms harness a unique cytotoxic strategy. In planarians, activin, a hormone regulating regeneration and reproduction, also acts as an inflammatory cytokine. Overactivation of activin signaling - through protein injection, genetic chimerism, or bacterial infection - triggers 'ruptoblasts', an undocumented immune cell type, to undergo 'ruptosis', a unique mode of cell bursting that eliminates nearby cells and bacteria in mere minutes, representing one of the fastest cytotoxic mechanisms observed. Ablating ruptoblasts suppresses inflammation but compromises bacterial clearance, highlighting ruptoblasts' broad-spectrum immune functions. We further identified ruptoblast-like cells in diverse basal bilaterians, unveiling an alternative strategy that couples hormonal regulation with immune defense and expanding the landscape of evolutionary immune innovations.
Collapse
|
9
|
Sujana STA, Shahjaman M, Singha AC. Application of bioinformatic tools in cell type classification for single-cell RNA-seq data. Comput Biol Chem 2025; 115:108332. [PMID: 39793515 DOI: 10.1016/j.compbiolchem.2024.108332] [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: 10/03/2024] [Revised: 12/06/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025]
Abstract
The advancements in single-cell RNA sequencing (scRNAseq) technology have significantly transformed genomics research, enabling the handling of thousands of cells in each experiment. As of now, 32,068 research studies have been cataloged in the Pubmed database. The primary aim of scRNAseq investigations is to identify cell types, understand the antitumor immune response, and identify new and uncommon cell types. Traditional techniques for identifying cell types include microscopy, histology, and pathological characteristics. However, the complexity of instruments and the need for precise experimental design make it difficult to fully capture the overall heterogeneity. Unsupervised clustering and supervised classification methods have been used to solve this task. Supervised cell type classification methods have gained popularity as large-scale, high-quality, well-annotated and more robust results compared to clustering methods. A recent study showed that support vector machine (SVM) gives a high-quality classification performance in different scenarios. In this article, we compare and evaluate the performance of four different kernels (sigmoid, linear, radial, polynomial) of SVM. The results of the experiments on three standard scRNA-seq datasets indicate that SVM with linear and SVM with sigmoid kernel classify the cells more accurately (approx. 99 %) where SVM linear kernel method has remarkably fast computation time and we also evaluate the results using some single cell specific evaluation matrices F-1 score, MCC, AUC value. Additionally, it sheds light on the potential use of kernels of SVM to give underlying information of single-cell RNA-Seq data more effectively.
Collapse
Affiliation(s)
- Shah Tania Akter Sujana
- Bioinformatics Lab, Department of Statistics, Begum Rokeya University, Rangpur 5404, Bangladesh.
| | - Md Shahjaman
- Bioinformatics Lab, Department of Statistics, Begum Rokeya University, Rangpur 5404, Bangladesh.
| | - Atul Chandra Singha
- Bioinformatics Lab, Department of Statistics, Begum Rokeya University, Rangpur 5404, Bangladesh.
| |
Collapse
|
10
|
Sun D, He S, Li X, Jin B, Wu F, Liu D, Dong Z, Chen G. Toxic effects and mechanistic insights of cadmium telluride quantum dots on the homeostasis and regeneration in planarians. JOURNAL OF HAZARDOUS MATERIALS 2025; 486:137047. [PMID: 39754879 DOI: 10.1016/j.jhazmat.2024.137047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
The widespread application of quantum dots (QDs) in recent years has raised concerns about potential environmental and human health risks. Although the toxicity of cadmium telluride quantum dots (CdTe QDs) has been partially studied, their effects on stem cells, tissue regeneration, neurodevelopment, and neurobehavioral toxicity remain unclear. This study aimed to investigate the combined toxic effects and mechanisms of CdTe QDs on planarians at the individual, tissue, cellular, and molecular levels. The results showed that exposure to CdTe QDs led to tissue damage, abnormal motor behavior, delayed regeneration, morphological abnormalities, and reduced survival. Furthermore, CdTe QDs caused excessive stem cell proliferation, leading to defective differentiation of tissues such as the epidermis, cilia, protonephridia, muscle, and nerves. Neurotoxicity manifests as a reduction in the number of neurons and neurotransmitter imbalance. Further studies revealed that CdTe QDs induced cell death by promoting reactive oxygen species (ROS) accumulation, triggering oxidative stress and deoxyribonucleic acid (DNA) damage, which led to excessive mitochondrial fission and activation of the mitochondria-dependent apoptotic signaling pathway. Overall, the balance between stem cell proliferation, differentiation, and apoptosis was disrupted, ultimately leading to delayed regeneration and homeostatic imbalance. These findings offer new insights into the environmental risk assessment of QDs and provide valuable directions for further research on their toxic effects on human stem cells and regenerative processes.
Collapse
Affiliation(s)
- Dandan Sun
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Siyuan He
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Xuheng Li
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Baijie Jin
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Fan Wu
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Dezeng Liu
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Zimei Dong
- College of Life Science, Henan Normal University, Xinxiang 453007, China.
| | - Guangwen Chen
- College of Life Science, Henan Normal University, Xinxiang 453007, China.
| |
Collapse
|
11
|
Verma P, Allen JM, Sánchez Alvarado A, Duncan EM. Chromatin remodeling protein BPTF mediates chromatin accessibility at gene promoters in planarian stem cells. BMC Genomics 2025; 26:232. [PMID: 40069606 PMCID: PMC11895202 DOI: 10.1186/s12864-025-11405-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The regulation of chromatin accessibility is essential in eukaryotic cells as one of several mechanisms that ensure gene activation occurs at appropriate times and in appropriate cell types. Accordingly, mutations in chromatin remodeling proteins are linked to many different developmental disorders and cancers. One example of a chromatin protein that has been linked to both developmental abnormalities and cancer is BPTF/NURF301, the largest subunit of the Nucleosome Remodeling Factor (NuRF) complex. The BPTF subunit is not only important for the formation of NuRF but also helps direct its activity to particular regions of chromatin by preferentially binding histone H3 lysine four trimethylation (H3K4me3). Notably, defects caused by knockdown of bptf in Xenopus embryos mimic those caused by knockdown of wdr5, a core subunit of all H3K4me3 methyltransferase complexes. However, the mechanistic details of how and where BPTF/NuRF is recruited to regulate gene expression vary between studies and have been largely tested in vitro and/or in cultured cells. Improving our understanding of how this chromatin remodeling complex targets specific gene loci and regulates their expression in an organismal context will provide important insight into how pathogenic mutations disrupt its normal, in vivo, cellular functions. RESULTS Here, we report our findings on the role of BPTF in maintaining chromatin accessibility and essential function in planarian (Schmidtea mediterranea) stem cells. We find that depletion of planarian BPTF primarily affects accessibility at gene promoters near transcription start sites (TSSs). BPTF-dependent loss of accessibility did not correlate with decreased gene expression when we considered all affected loci. However, we found that genes marked by Set1-dependent H3K4me3, but not MLL1/2-dependent H3K4me3, showed increased sensitivity to the loss of BPTF-dependent accessibility. In addition, knockdown of bptf (Smed-bptf) produces loss-of-function phenotypes similar to those caused by knockdown of Smed-set1. CONCLUSIONS The S.mediterranea homolog of NuRF protein BPTF (SMED-BPTF) is essential for normal homeostasis in planarian tissues, potentially through its role in maintaining chromatin accessibility at a specific subset of gene promoters in planarian stem cells. By identifying loci that lose both chromatin accessibility and gene expression after depletion of BPTF, we have identified a cohort of genes that may have important functions in stem cell biology.
Collapse
Affiliation(s)
- Prince Verma
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - John M Allen
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | | | | |
Collapse
|
12
|
Lo KC, Petersen CP. map3k1 suppresses terminal differentiation of migratory eye progenitors in planarian regeneration. PLoS Genet 2025; 21:e1011457. [PMID: 40096024 PMCID: PMC11981174 DOI: 10.1371/journal.pgen.1011457] [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: 10/09/2024] [Revised: 04/09/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025] Open
Abstract
Proper stem cell targeting and differentiation is necessary for regeneration to succeed. In organisms capable of whole body regeneration, considerable progress has been made identifying wound signals initiating this process, but the mechanisms that control the differentiation of progenitors into mature organs are not fully understood. Using the planarian as a model system, we identify a novel function for map3k1, a MAP3K family member possessing both kinase and ubiquitin ligase domains, to negatively regulate terminal differentiation of stem cells during eye regeneration. Inhibition of map3k1 caused the formation of multiple ectopic eyes within the head, but without controlling overall head, brain, or body patterning. By contrast, other known regulators of planarian eye patterning like wnt11-6/wntA and notum also regulate head regionalization, suggesting map3k1 acts distinctly. Consistent with these results, eye resection and regeneration experiments suggest that unlike Wnt signaling perturbation, map3k1 inhibition did not shift the target destination of eye formation in the animal. map3k1(RNAi) ectopic eyes emerged in the regions normally occupied by migratory eye progenitors, and these animals produced a net excess of differentiated eye cells. Furthermore, the formation of ectopic eyes after map3k1 inhibition coincided with an increase to numbers of differentiated eye cells, a decrease in numbers of ovo+ eye progenitors, and also was preceded by eye progenitors prematurely expressing opsin/tyosinase markers of eye cell terminal differentiation. Therefore, map3k1 negatively regulates the process of terminal differentiation within the eye lineage. Similar ectopic eye phenotypes were also observed after inhibition of map2k4, map2k7, jnk, and p38, identifying a putative pathway through which map3k1 prevents differentiation. Together, these results suggest that map3k1 regulates a novel control point in the eye regeneration pathway which suppresses the terminal differentiation of progenitors during their migration to target destinations.
Collapse
Affiliation(s)
- Katherine C. Lo
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| | - Christian P. Petersen
- Department of Molecular Biosciences, Northwestern University, Evanston, Illinois, United States of America
| |
Collapse
|
13
|
Goldman OV, DeFoe AE, Qi Y, Jiao Y, Weng SC, Houri-Zeevi L, Lakhiani P, Morita T, Razzauti J, Rosas-Villegas A, Tsitohay YN, Walker MM, Hopkins BR, Akbari OS, Duvall LB, White-Cooper H, Sorrells TR, Sharma R, Li H, Vosshall LB, Shai N. Mosquito Cell Atlas: A single-nucleus transcriptomic atlas of the adult Aedes aegypti mosquito. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.639765. [PMID: 40060408 PMCID: PMC11888250 DOI: 10.1101/2025.02.25.639765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The female mosquito's remarkable ability to hunt humans and transmit pathogens relies on her unique biology. Here, we present the Mosquito Cell Atlas (MCA), a comprehensive single-nucleus RNA sequencing dataset of more than 367,000 nuclei from 19 dissected tissues of adult female and male Aedes aegypti, providing cellular-level resolution of mosquito biology. We identify novel cell types and expand our understanding of sensory neuron organization of chemoreceptors to all sensory tissues. Our analysis uncovers male-specific cells and sexually dimorphic gene expression in the antenna and brain. In female mosquitoes, we find that glial cells in the brain, rather than neurons, undergo the most extensive transcriptional changes following blood feeding. Our findings provide insights into the cellular basis of mosquito behavior and sexual dimorphism. The MCA aims to serve as a resource for the vector biology community, enabling systematic investigation of cell-type specific expression across all mosquito tissues.
Collapse
Affiliation(s)
- Olivia V. Goldman
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
| | - Alexandra E. DeFoe
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yaoyu Jiao
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Shih-Che Weng
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Leah Houri-Zeevi
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Priyanka Lakhiani
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Takeshi Morita
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Jacopo Razzauti
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Price Family Center for the Social Brain, The Rockefeller University, New York, NY 10065, USA
| | - Adriana Rosas-Villegas
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Yael N. Tsitohay
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
| | - Madison M. Walker
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Ben R. Hopkins
- Department of Evolution and Ecology, University of California Davis, Davis, CA 95616, USA
| | | | - Omar S. Akbari
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laura B. Duvall
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Helen White-Cooper
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Trevor R. Sorrells
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Wu Tsai Institute, Yale University, New Haven, CT 06510, USA
- Howard Hughes Medical Institute, New Haven, CT 06510, USA
| | - Roshan Sharma
- Program for Computational and Systems Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Single-cell Analytics Innovation Lab, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leslie B. Vosshall
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Kavli Neural Systems Institute, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Nadav Shai
- Laboratory of Neurogenetics and Behavior, The Rockefeller University, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| |
Collapse
|
14
|
Kuznetsov NV, Statsenko Y, Ljubisavljevic M. An Update on Neuroaging on Earth and in Spaceflight. Int J Mol Sci 2025; 26:1738. [PMID: 40004201 PMCID: PMC11855577 DOI: 10.3390/ijms26041738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Over 400 articles on the pathophysiology of brain aging, neuroaging, and neurodegeneration were reviewed, with a focus on epigenetic mechanisms and numerous non-coding RNAs. In particular, this review the accent is on microRNAs, the discovery of whose pivotal role in gene regulation was recognized by the 2024 Nobel Prize in Physiology or Medicine. Aging is not a gradual process that can be easily modeled and described. Instead, multiple temporal processes occur during aging, and they can lead to mosaic changes that are not uniform in pace. The rate of change depends on a combination of external and internal factors and can be boosted in accelerated aging. The rate can decrease in decelerated aging due to individual structural and functional reserves created by cognitive, physical training, or pharmacological interventions. Neuroaging can be caused by genetic changes, epigenetic modifications, oxidative stress, inflammation, lifestyle, and environmental factors, which are especially noticeable in space environments where adaptive changes can trigger aging-like processes. Numerous candidate molecular biomarkers specific to neuroaging need to be validated to develop diagnostics and countermeasures.
Collapse
Affiliation(s)
- Nik V. Kuznetsov
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
| | - Yauhen Statsenko
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Radiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Milos Ljubisavljevic
- ASPIRE Precision Medicine Research Institute Abu Dhabi, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (Y.S.); (M.L.)
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
15
|
Moescheid MF, Lu Z, Soria CD, Quack T, Puckelwaldt O, Holroyd N, Holzaepfel P, Haeberlein S, Rinaldi G, Berriman M, Grevelding CG. The retinoic acid family-like nuclear receptor SmRAR identified by single-cell transcriptomics of ovarian cells controls oocyte differentiation in Schistosoma mansoni. Nucleic Acids Res 2025; 53:gkae1228. [PMID: 39676663 PMCID: PMC11879061 DOI: 10.1093/nar/gkae1228] [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: 04/09/2024] [Revised: 10/30/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024] Open
Abstract
Studies on transcription regulation in platyhelminth development are scarce, especially for parasitic flatworms. Here, we employed single-cell transcriptomics to identify genes involved in reproductive development in the trematode model Schistosoma mansoni. This parasite causes schistosomiasis, a major neglected infectious disease affecting >240 million people worldwide. The pathology of schistosomiasis is closely associated with schistosome eggs deposited in host organs including the liver. Unlike other trematodes, schistosomes exhibit distinct sexes, with egg production reliant on the pairing-dependent maturation of female reproductive organs. Despite this significance, the molecular mechanisms underlying ovary development and oocyte differentiation remain largely unexplored. Utilizing an organ isolation approach for S. mansoni, we extracted ovaries of paired females followed by single-cell RNA sequencing (RNA-seq) with disassociated oocytes. A total of 1967 oocytes expressing 7872 genes passed quality control (QC) filtering. Unsupervised clustering revealed four distinct cell clusters: somatic, germ cells and progeny, intermediate and late germ cells. Among distinct marker genes for each cluster, we identified a hitherto uncharacterized transcription factor of the retinoic acid receptor family, SmRAR. Functional analyses of SmRAR and associated genes like Smmeiob (meiosis-specific, oligonucleotide/oligosaccharide binding motif (OB) domain-containing) demonstrated their pairing-dependent and ovary-preferential expression and their decisive roles in oocyte differentiation of S. mansoni.
Collapse
Affiliation(s)
- Max F Moescheid
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Zhigang Lu
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Carmen Diaz Soria
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Thomas Quack
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Oliver Puckelwaldt
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Nancy Holroyd
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
| | - Pauline Holzaepfel
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Simone Haeberlein
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| | - Gabriel Rinaldi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- Department of Life Sciences, Aberystwyth University, Penglais, Aberystwyth, Ceredigion, SY23 3DA, UK
| | - Matthew Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10, 1SA, UK
- School of Infection and Immunity, College of Medicine, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Christoph G Grevelding
- Institute of Parasitology, Justus Liebig University, Schubertstrasse 81, 35392 Giessen, Germany
| |
Collapse
|
16
|
Gąsiorowski L, Chai C, Rozanski A, Purandare G, Ficze F, Mizi A, Wang B, Rink JC. Regeneration in the absence of canonical neoblasts in an early branching flatworm. Nat Commun 2025; 16:1232. [PMID: 39890822 PMCID: PMC11785736 DOI: 10.1038/s41467-024-54716-x] [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: 06/10/2024] [Accepted: 11/19/2024] [Indexed: 02/03/2025] Open
Abstract
The remarkable regenerative abilities of flatworms are closely linked to neoblasts - adult pluripotent stem cells that are the only division-competent cell type outside of the reproductive system. Although the presence of neoblast-like cells and whole-body regeneration in other animals has led to the idea that these features may represent the ancestral metazoan state, the evolutionary origin of both remains unclear. Here we show that the catenulid Stenostomum brevipharyngium, a member of the earliest-branching flatworm lineage, lacks conventional neoblasts despite being capable of whole-body regeneration and asexual reproduction. Using a combination of single-nuclei transcriptomics, in situ gene expression analysis, and functional experiments, we find that cell divisions are not restricted to a single cell type and are associated with multiple fully differentiated somatic tissues. Furthermore, the cohort of germline multipotency genes, which are considered canonical neoblast markers, are not expressed in dividing cells, but in the germline instead, and we experimentally show that they are neither necessary for proliferation nor regeneration. Overall, our results challenge the notion that canonical neoblasts are necessary for flatworm regeneration and open up the possibility that neoblast-like cells may have evolved convergently in different animals, independent of their regenerative capacity.
Collapse
Affiliation(s)
- Ludwik Gąsiorowski
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Andrei Rozanski
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gargi Purandare
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Fruzsina Ficze
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Athanasia Mizi
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
17
|
Liu J, Yang M, Yu Y, Xu H, Wang T, Li K, Zhou X. Advancing bioinformatics with large language models: components, applications and perspectives. ARXIV 2025:arXiv:2401.04155v2. [PMID: 38259343 PMCID: PMC10802675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Large language models (LLMs) are a class of artificial intelligence models based on deep learning, which have great performance in various tasks, especially in natural language processing (NLP). Large language models typically consist of artificial neural networks with numerous parameters, trained on large amounts of unlabeled input using self-supervised or semi-supervised learning. However, their potential for solving bioinformatics problems may even exceed their proficiency in modeling human language. In this review, we will provide a comprehensive overview of the essential components of large language models (LLMs) in bioinformatics, spanning genomics, transcriptomics, proteomics, drug discovery, and single-cell analysis. Key aspects covered include tokenization methods for diverse data types, the architecture of transformer models, the core attention mechanism, and the pre-training processes underlying these models. Additionally, we will introduce currently available foundation models and highlight their downstream applications across various bioinformatics domains. Finally, drawing from our experience, we will offer practical guidance for both LLM users and developers, emphasizing strategies to optimize their use and foster further innovation in the field.
Collapse
Affiliation(s)
- Jiajia Liu
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, 77030, USA
| | - Mengyuan Yang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yankai Yu
- School of Computing and Artificial Intelligence, Southwest Jiaotong University, Chengdu, Sichuan 611756, China
| | - Haixia Xu
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, 77030, USA
| | - Tiangang Wang
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, 77030, USA
| | - Kang Li
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas, 77030, USA
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
18
|
Chen X. Stem cells (neoblasts) and positional information jointly dominate regeneration in planarians. Heliyon 2025; 11:e41833. [PMID: 39877626 PMCID: PMC11773080 DOI: 10.1016/j.heliyon.2025.e41833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/18/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
Regeneration is the ability to accurately regrow missing body parts. The unparalleled regenerative capacity and incredible tissue plasticity of planarians, both resulting from the presence of abundant adult stem cells referred to as neoblasts, offer a unique opportunity to investigate the cellular and molecular principles underlying regeneration. Neoblasts are capable of self-renewal and differentiation into the desired cell types for correct replacement of lost parts after tissue damage. Positional information in muscle cells governs the polarity and patterning of the body plan during homeostasis and regeneration. For planarians, removal of neoblasts disables the regenerative feats and disruption of positional information results in the regeneration of inappropriate missing body regions, only the combination of neoblasts and positional information enables regeneration. Here, I summarize the current state of the field in neoblast lineage potential, subclasses and specification, and in the roles of positional information for proper tissue turnover and regeneration in planarians.
Collapse
Affiliation(s)
- Xuhui Chen
- Affiliated Infectious Diseases Hospital of Zhengzhou University (Henan Infectious Diseases Hospital, The Sixth People's Hospital of Zhengzhou), Center for Translational Medicine, Zhengzhou, 45000, China
| |
Collapse
|
19
|
Zhong H, Han W, Gomez-Cabrero D, Tegner J, Gao X, Cui G, Aranda M. Benchmarking cross-species single-cell RNA-seq data integration methods: towards a cell type tree of life. Nucleic Acids Res 2025; 53:gkae1316. [PMID: 39778870 PMCID: PMC11707536 DOI: 10.1093/nar/gkae1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/23/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Cross-species single-cell RNA-seq data hold immense potential for unraveling cell type evolution and transferring knowledge between well-explored and less-studied species. However, challenges arise from interspecific genetic variation, batch effects stemming from experimental discrepancies and inherent individual biological differences. Here, we benchmarked nine data-integration methods across 20 species, encompassing 4.7 million cells, spanning eight phyla and the entire animal taxonomic hierarchy. Our evaluation reveals notable differences between the methods in removing batch effects and preserving biological variance across taxonomic distances. Methods that effectively leverage gene sequence information capture underlying biological variances, while generative model-based approaches excel in batch effect removal. SATURN demonstrates robust performance across diverse taxonomic levels, from cross-genus to cross-phylum, emphasizing its versatility. SAMap excels in integrating species beyond the cross-family level, especially for atlas-level cross-species integration, while scGen shines within or below the cross-class hierarchy. As a result, our analysis offers recommendations and guidelines for selecting suitable integration methods, enhancing cross-species single-cell RNA-seq analyses and advancing algorithm development.
Collapse
Affiliation(s)
- Huawen Zhong
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Wenkai Han
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David Gomez-Cabrero
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Translational Bioinformatics, Navarrabiomed—Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Jesper Tegner
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, SE-171 76 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, SE-17165 Solna, Sweden
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Guoxin Cui
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Manuel Aranda
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
20
|
Scimone ML, Canales BII, Aoude P, Atabay KD, Reddien PW. Coordinated neuron-glia regeneration through Notch signaling in planarians. PLoS Genet 2025; 21:e1011577. [PMID: 39869602 PMCID: PMC11801701 DOI: 10.1371/journal.pgen.1011577] [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: 08/21/2024] [Revised: 02/06/2025] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Some animals can regenerate large missing regions of their nervous system, requiring mechanisms to restore the pattern, numbers, and wiring of diverse neuron classes. Because injuries are unpredictable, regeneration must be accomplished from an unlimited number of starting points. Coordinated regeneration of neuron-glia architecture is thus a major challenge and remains poorly understood. In planarians, neurons and glia are regenerated from distinct progenitors. We found that planarians first regenerate neurons expressing a Delta-encoding gene, delta-2, at key positions in the central and peripheral nervous systems. Planarian glia are specified later from dispersed Notch-1-expressing mesoderm-like phagocytic progenitors. Inhibition of delta-2 or notch-1 severely reduced glia in planarians, but did not affect the specification of other phagocytic cell types. Loss of several delta-2-expressing neuron classes prevented differentiation of the glia associated with them, whereas transplantation of delta-2-expressing photoreceptor neurons was sufficient for glia formation at an ectopic location. Our results suggest a model in which patterned delta-2-expressing neurons instruct phagocytic progenitors to locally differentiate into glia, presenting a mechanism for coordinated regeneration of numbers and pattern of cell types.
Collapse
Affiliation(s)
- M. Lucila Scimone
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Bryanna Isela-Inez Canales
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Patrick Aoude
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
- Department of Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Kutay D. Atabay
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Peter W. Reddien
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| |
Collapse
|
21
|
Pan X, Zhao Y, Li Y, Chen J, Zhang W, Yang L, Xiong YZ, Ying Y, Xu H, Zhang Y, Gao C, Sun Y, Li N, Chen L, Chen Z, Lei K. Mitochondrial dynamics govern whole-body regeneration through stem cell pluripotency and mitonuclear balance. Nat Commun 2024; 15:10681. [PMID: 39672898 PMCID: PMC11645412 DOI: 10.1038/s41467-024-54720-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 11/19/2024] [Indexed: 12/15/2024] Open
Abstract
Tissue regeneration is a complex process involving large changes in cell proliferation, fate determination, and differentiation. Mitochondrial dynamics and metabolism play a crucial role in development and wound repair, but their function in large-scale regeneration remains poorly understood. Planarians offer an excellent model to investigate this process due to their remarkable regenerative abilities. In this study, we examine mitochondrial dynamics during planarian regeneration. We find that knockdown of the mitochondrial fusion gene, opa1, impairs both tissue regeneration and stem cell pluripotency. Interestingly, the regeneration defects caused by opa1 knockdown are rescued by simultaneous knockdown of the mitochondrial fission gene, drp1, which partially restores mitochondrial dynamics. Furthermore, we discover that Mitolow stem cells exhibit an enrichment of pluripotency due to their fate choices at earlier stages. Transcriptomic analysis reveals the delicate mitonuclear balance in metabolism and mitochondrial proteins in regeneration, controlled by mitochondrial dynamics. These findings highlight the importance of maintaining mitochondrial dynamics in large-scale tissue regeneration and suggest the potential for manipulating these dynamics to enhance stem cell functionality and regenerative processes.
Collapse
Affiliation(s)
- Xue Pan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yun Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Yucong Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Jiajia Chen
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wenya Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Ling Yang
- HPC Center, Westlake University, Hangzhou, Zhejiang, China
| | - Yuanyi Zhou Xiong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Yuqing Ying
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Hao Xu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuhong Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Chong Gao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yuhan Sun
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Nan Li
- HPC Center, Westlake University, Hangzhou, Zhejiang, China
| | - Liangyi Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, National Biomedical Imaging Center, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
- State Key Laboratory of Membrane Biology, Peking University, Beijing, China.
- PKU-Nanjing Institute of Translational Medicine, Nanjing, China.
| | - Zhixing Chen
- College of Future Technology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, National Biomedical Imaging Center, Peking University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| | - Kai Lei
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
22
|
Chen J, Li Y, Wang Y, Wang H, Yang J, Pan X, Zhao Y, Xu H, Jiang P, Qian P, Wang H, Xie Z, Lei K. Fibrillarin homologs regulate translation in divergent cell lineages during planarian homeostasis and regeneration. EMBO J 2024; 43:6591-6625. [PMID: 39567829 PMCID: PMC11649923 DOI: 10.1038/s44318-024-00315-x] [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: 08/08/2022] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Tissue homeostasis and regeneration involve complex cellular changes. The role of rRNA modification-dependent translational regulation in these processes remains largely unknown. Planarians, renowned for their ability to undergo remarkable tissue regeneration, provide an ideal model for the analysis of differential rRNA regulation in diverse cell types during tissue homeostasis and regeneration. We investigated the role of RNA 2'-O-methyltransferase, Fibrillarin (FBL), in the planarian Schmidtea mediterranea and identified two FBL homologs: Smed-fbl-1 (fbl-1) and Smed-fbl-2 (fbl-2). Both are essential for planarian regeneration, but play distinct roles: fbl-1 is crucial for progenitor cell differentiation, while fbl-2 is important for late-stage epidermal lineage specification. Different 2'-O-methylation patterns were observed upon fbl-1 and fbl-2 knockdown, suggesting their roles in translation of specific mRNA pools during regeneration. Ribo-seq analysis further revealed differing impacts of fbl-1 and fbl-2 knockdown on gene translation. These findings indicate divergent roles of the duplicate fbl genes in specific cell lineage development in planarians and suggest a role of rRNA modifications in translational regulation during tissue maintenance and regeneration.
Collapse
Affiliation(s)
- Jiajia Chen
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yucong Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Yan Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Vision Science, Guangzhou, China
| | - Hui Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Jiaqi Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Vision Science, Guangzhou, China
| | - Xue Pan
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yun Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Hao Xu
- School of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Penglei Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University & Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Hongwei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Vision Science, Guangzhou, China
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Vision Science, Guangzhou, China
| | - Kai Lei
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Lall S, Ray S, Bandyopadhyay S. Enhancing Single-Cell RNA-seq Data Completeness with a Graph Learning Framework. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; PP:64-72. [PMID: 39504287 DOI: 10.1109/tcbb.2024.3492384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Single cell RNA sequencing (scRNA-seq) is a powerful tool to capture gene expression snapshots in individual cells. However, a low amount of RNA in the individual cells results in dropout events, which introduce huge zero counts in the single cell expression matrix. We have developed VAImpute, a variational graph autoencoder based imputation technique that learns the inherent distribution of a large network/graph constructed from the scRNA-seq data leveraging copula correlation () among cells/genes. The trained model is utilized to predict the dropouts events by computing the probability of all non-edges (cell-gene) in the network. We devise an algorithm to impute the missing expression values of the detected dropouts. The performance of the proposed model is assessed on both simulated and real scRNA-seq datasets, comparing it to established single-cell imputation methods. VAImpute yields significant improvements to detect dropouts, thereby achieving superior performance in cell clustering, detecting rare cells, and differential expression. All codes and datasets are given in the github link: https://github.com/sumantaray/VAImputeAvailability.
Collapse
|
24
|
Zeng Y, Ma Q, Chen J, Kong X, Chen Z, Liu H, Liu L, Qian Y, Wang X, Lu S. Single-cell sequencing: Current applications in various tuberculosis specimen types. Cell Prolif 2024; 57:e13698. [PMID: 38956399 PMCID: PMC11533074 DOI: 10.1111/cpr.13698] [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: 01/24/2024] [Revised: 05/21/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Tuberculosis (TB) is a chronic disease caused by Mycobacterium tuberculosis (M.tb) and responsible for millions of deaths worldwide each year. It has a complex pathogenesis that primarily affects the lungs but can also impact systemic organs. In recent years, single-cell sequencing technology has been utilized to characterize the composition and proportion of immune cell subpopulations associated with the pathogenesis of TB disease since it has a high resolution that surpasses conventional techniques. This paper reviews the current use of single-cell sequencing technologies in TB research and their application in analysing specimens from various sources of TB, primarily peripheral blood and lung specimens. The focus is on how these technologies can reveal dynamic changes in immune cell subpopulations, genes and proteins during disease progression after M.tb infection. Based on the current findings, single-cell sequencing has significant potential clinical value in the field of TB research. Next, we will focus on the real-world applications of the potential targets identified through single-cell sequencing for diagnostics, therapeutics and the development of effective vaccines.
Collapse
Affiliation(s)
- Yuqin Zeng
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Quan Ma
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Jinyun Chen
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Xingxing Kong
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Zhanpeng Chen
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Huazhen Liu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Lanlan Liu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Yan Qian
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Xiaomin Wang
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| | - Shuihua Lu
- National Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalShenzhenGuangdong ProvinceChina
| |
Collapse
|
25
|
Medlock-Lanier T, Clay KB, Roberts-Galbraith RH. Planarian LDB and SSDP proteins scaffold transcriptional complexes for regeneration and patterning. Dev Biol 2024; 515:67-78. [PMID: 38968988 PMCID: PMC11361279 DOI: 10.1016/j.ydbio.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
Sequence-specific transcription factors often function as components of large regulatory complexes. LIM-domain binding protein (LDB) and single-stranded DNA-binding protein (SSDP) function as core scaffolds of transcriptional complexes in animals and plants. Little is known about potential partners and functions for LDB/SSDP complexes in the context of tissue regeneration. In this work, we find that planarian LDB1 and SSDP2 promote tissue regeneration, with a particular function in anterior regeneration and mediolateral polarity reestablishment. We find that LDB1 and SSDP2 interact with one another and with characterized planarian LIM-HD proteins Arrowhead, Islet1, and Lhx1/5-1. We also show that SSDP2 and LDB1 function with islet1 in polarity reestablishment and with lhx1/5-1 in serotonergic neuron maturation. Finally, we find new roles for LDB1 and SSDP2 in regulating gene expression in the planarian intestine and parenchyma; these functions are likely LIM-HD-independent. Together, our work provides insight into LDB/SSDP complexes in a highly regenerative organism. Further, our work provides a strong starting point for identifying and characterizing potential binding partners of LDB1 and SSDP2 and for exploring roles for these proteins in diverse aspects of planarian physiology.
Collapse
Affiliation(s)
| | - Kendall B Clay
- Neuroscience Program, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
26
|
Lo KC, Petersen CP. map3k1 suppresses terminal differentiation of migratory eye progenitors in planarian regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617745. [PMID: 39416008 PMCID: PMC11483071 DOI: 10.1101/2024.10.11.617745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Proper stem cell targeting and differentiation is necessary for regeneration to succeed. In organisms capable of whole body regeneration, considerable progress has been made identifying wound signals initiating this process, but the mechanisms that control the differentiation of progenitors into mature organs are not fully understood. Using the planarian as a model system, we identify a novel function for map3k1, a MAP3K family member possessing both kinase and ubiquitin ligase domains, to negatively regulate terminal differentiation of stem cells during eye regeneration. Inhibition of map3k1 caused the formation of multiple ectopic eyes within the head, but without controlling overall head, brain, or body patterning. By contrast, other known regulators of planarian eye patterning like WntA and notum also regulate head regionalization, suggesting map3k1 acts distinctly. Eye resection and regeneration experiments suggest that unlike Wnt signaling perturbation, map3k1 inhibition did not shift the target destination of eye formation in the animal. Instead, map3k1(RNAi) ectopic eyes emerge in the regions normally occupied by migratory eye progenitors, and the onset of ectopic eyes after map3k1 inhibition coincides with a reduction to eye progenitor numbers. Furthermore, RNAi dosing experiments indicate that progenitors closer to their normal target are relatively more sensitive to the effects of map3k1, implicating this factors in controlling the site of terminal differentiation. Eye phenotypes were also observed after inhibition of map2k4, map2k7, jnk, and p38, identifying a putative pathway through which map3k1 prevents differentiation. Together, these results suggest that map3k1 regulates a novel control point in the eye regeneration pathway which suppresses the terminal differentiation of progenitors during their migration to target destinations.
Collapse
Affiliation(s)
- Katherine C. Lo
- Department of Molecular Biosciences, Northwestern University
| | | |
Collapse
|
27
|
Xing N, Gao L, Xie W, Deng H, Yang F, Liu D, Li A, Pang Q. Mining of potentially stem cell-related miRNAs in planarians. Mol Biol Rep 2024; 51:1045. [PMID: 39377855 DOI: 10.1007/s11033-024-09977-6] [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: 03/20/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
Stem cells and regenerative medicine have recently become important research topics. However, the complex stem cell regulatory networks involved in various microRNA (miRNA)-mediated mechanisms have not yet been fully elucidated. Planarians are ideal animal models for studying stem cells owing to their rich stem cell populations (neoblasts) and extremely strong regeneration capacity. The roles of planarian miRNAs in stem cells and regeneration have long attracted attention. However, previous studies have generally provided simple datasets lacking integrative analysis. Here, we have summarized the miRNA family reported in planarians and highlighted conservation in both sequence and function. Furthermore, we summarized miRNA data related to planarian stem cells and regeneration and screened potential involved candidates. Nevertheless, the roles of these miRNAs in planarian regeneration and stem cells remain unclear. The identification of potential stem cell-related miRNAs offers more precise suggestions and references for future investigations of miRNAs in planarians. Furthermore, it provides potential research avenues for understanding the mechanisms of stem cell regulatory networks. Finally, we compiled a summary of the experimental methods employed for studying planarian miRNAs, with the aim of highlighting special considerations in certain procedures and providing more convenient technical support for future research endeavors.
Collapse
Affiliation(s)
- Nianhong Xing
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Lili Gao
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China.
| | - Wenshuo Xie
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Hongkuan Deng
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Fengtang Yang
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Dongwu Liu
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Ao Li
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institute, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, 255049, China.
| |
Collapse
|
28
|
Mukundan N, Hariharan N, Sasidharan V, Lakshmanan V, Palakodeti D, Jamora C. Poly (A) binding protein 2 is critical for stem cell differentiation during regeneration in the planarian Schmidtea mediterranea. Front Cell Dev Biol 2024; 12:1433142. [PMID: 39376632 PMCID: PMC11456742 DOI: 10.3389/fcell.2024.1433142] [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: 05/15/2024] [Accepted: 08/21/2024] [Indexed: 10/09/2024] Open
Abstract
Post-transcriptional regulation has emerged as a key mechanism for regulating stem cell renewal and differentiation, which is essential for understanding tissue regeneration and homeostasis. Poly(A)-binding proteins are a family of RNA-binding proteins that play a vital role in post-transcriptional regulation by controlling mRNA stability and protein synthesis. The involvement of poly(A) binding proteins in a wide range of cellular functions is increasingly being investigated. In this study, we used the regenerative model planarian organism Schmidtea mediterranea to demonstrate the critical role of poly(A)-binding protein 2 (PABP2) in regulating neoblast maintenance and differentiation. A deficit in PABP2 blocks the transition of neoblasts toward immediate early progenitors, leading to an enhanced pool of non-committed neoblasts and a decreased progenitor population. This is reflected in variations in the transcriptome profile, providing evidence of downregulation in multiple lineages. Thus, an insufficiency of PABP2 resulted in defective formation and organization of tissue, leading to abnormal regeneration. Our study reveals the essential role of PABP2 in regulating genes that mediate stem cell commitment to early progenitors during tissue regeneration.
Collapse
Affiliation(s)
- Namita Mukundan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nivedita Hariharan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | | | - Vairavan Lakshmanan
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Dasaradhi Palakodeti
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Colin Jamora
- Integrative Chemical Biology (ICB), Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
- Department of Life Science, Shiv Nadar Institution of Eminence, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
29
|
Ivanković M, Brand JN, Pandolfini L, Brown T, Pippel M, Rozanski A, Schubert T, Grohme MA, Winkler S, Robledillo L, Zhang M, Codino A, Gustincich S, Vila-Farré M, Zhang S, Papantonis A, Marques A, Rink JC. A comparative analysis of planarian genomes reveals regulatory conservation in the face of rapid structural divergence. Nat Commun 2024; 15:8215. [PMID: 39294119 PMCID: PMC11410931 DOI: 10.1038/s41467-024-52380-9] [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: 01/20/2024] [Accepted: 08/30/2024] [Indexed: 09/20/2024] Open
Abstract
The planarian Schmidtea mediterranea is being studied as a model species for regeneration, but the assembly of planarian genomes remains challenging. Here, we report a high-quality haplotype-phased, chromosome-scale genome assembly of the sexual S2 strain of S. mediterranea and high-quality chromosome-scale assemblies of its three close relatives, S. polychroa, S. nova, and S. lugubris. Using hybrid gene annotations and optimized ATAC-seq and ChIP-seq protocols for regulatory element annotation, we provide valuable genome resources for the planarian research community and a first comparative perspective on planarian genome evolution. Our analyses reveal substantial divergence in protein-coding sequences and regulatory regions but considerable conservation within promoter and enhancer annotations. We also find frequent retrotransposon-associated chromosomal inversions and interchromosomal translocations within the genus Schmidtea and, remarkably, independent and nearly complete losses of ancestral metazoan synteny in Schmidtea and two other flatworm groups. Overall, our results suggest that platyhelminth genomes can evolve without syntenic constraints.
Collapse
Affiliation(s)
- Mario Ivanković
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jeremias N Brand
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Luca Pandolfini
- Center for Human Technologies, Non-coding RNA and RNA-based therapeutics, Istituto Italiano di Tecnologia, Genova, Italy
| | - Thomas Brown
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Martin Pippel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andrei Rozanski
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Til Schubert
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Markus A Grohme
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Laura Robledillo
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Meng Zhang
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Azzurra Codino
- Center for Human Technologies, Non-coding RNA and RNA-based therapeutics, Istituto Italiano di Tecnologia, Genova, Italy
| | - Stefano Gustincich
- Center for Human Technologies, Non-coding RNA and RNA-based therapeutics, Istituto Italiano di Tecnologia, Genova, Italy
| | - Miquel Vila-Farré
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shu Zhang
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Argyris Papantonis
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - André Marques
- Department of Chromosome Biology, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Faculty of Biology und Psychology, Georg-August-University Göttingen, Göttingen, Germany.
| |
Collapse
|
30
|
Ross KG, Alvarez Zepeda S, Auwal MA, Garces AK, Roman S, Zayas RM. The Role of Polycystic Kidney Disease-Like Homologs in Planarian Nervous System Regeneration and Function. Integr Org Biol 2024; 6:obae035. [PMID: 39364443 PMCID: PMC11448475 DOI: 10.1093/iob/obae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/05/2024] Open
Abstract
Planarians are an excellent model for investigating molecular mechanisms necessary for regenerating a functional nervous system. Numerous studies have led to the generation of extensive genomic resources, especially whole-animal single-cell RNA-seq resources. These have facilitated in silico predictions of neuronal subtypes, many of which have been anatomically mapped by in situ hybridization. However, our knowledge of the function of dozens of neuronal subtypes remains poorly understood. Previous investigations identified that polycystic kidney disease (pkd)-like genes in planarians are strongly expressed in sensory neurons and have roles in mechanosensation. Here, we examine the expression and function of all the pkd genes found in the Schmidtea mediterranea genome and map their expression in the asexual and hermaphroditic strains. Using custom behavioral assays, we test the function of pkd genes in response to mechanical stimulation and in food detection. Our work provides insight into the physiological function of sensory neuron populations and protocols for creating inexpensive automated setups for acquiring and analyzing mechanosensory stimulation in planarians.
Collapse
Affiliation(s)
- K G Ross
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - S Alvarez Zepeda
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - M A Auwal
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - A K Garces
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - S Roman
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| | - R M Zayas
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182, USA
| |
Collapse
|
31
|
Chai C, Gibson J, Li P, Pampari A, Patel A, Kundaje A, Wang B. Flexible use of conserved motif vocabularies constrains genome access in cell type evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611027. [PMID: 39282369 PMCID: PMC11398382 DOI: 10.1101/2024.09.03.611027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Cell types evolve into a hierarchy with related types grouped into families. How cell type diversification is constrained by the stable separation between families over vast evolutionary times remains unknown. Here, integrating single-nucleus multiomic sequencing and deep learning, we show that hundreds of sequence features (motifs) divide into distinct sets associated with accessible genomes of specific cell type families. This division is conserved across highly divergent, early-branching animals including flatworms and cnidarians. While specific interactions between motifs delineate cell type relationships within families, surprisingly, these interactions are not conserved between species. Consistently, while deep learning models trained on one species can predict accessibility of other species' sequences, their predictions frequently rely on distinct, but synonymous, motif combinations. We propose that long-term stability of cell type families is maintained through genome access specified by conserved motif sets, or 'vocabularies', whereas cell types diversify through flexible use of motifs within each set.
Collapse
Affiliation(s)
- Chew Chai
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Jesse Gibson
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Pengyang Li
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Anusri Pampari
- Department of Computer Science, Stanford University, Stanford, USA
| | - Aman Patel
- Department of Computer Science, Stanford University, Stanford, USA
| | - Anshul Kundaje
- Department of Computer Science, Stanford University, Stanford, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, USA
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
32
|
Curry HN, Huynh R, Rouhana L. Melastatin subfamily Transient Receptor Potential channels support spermatogenesis in planarian flatworms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.01.610670. [PMID: 39282438 PMCID: PMC11398416 DOI: 10.1101/2024.09.01.610670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The Transient Receptor Potential superfamily of proteins (TRPs) form cation channels that are abundant in animal sensory systems. Amongst TRPs, the Melastatin-related subfamily (TRPMs) is composed of members that respond to temperature, pH, sex hormones, and various other stimuli. Some TRPMs exhibit enriched expression in gonads of vertebrate and invertebrate species, but their contributions to germline development remain to be determined. We identified twenty-one potential TRPMs in the planarian flatworm Schmidtea mediterranea and analyzed their anatomical distribution of expression by whole-mount in situ hybridization. Enriched expression of two TRPMs (Smed-TRPM-c and Smed-TRPM-l) was detected in testis, whereas eight TRPM genes had detectable expression in patterns representative of neuronal and/or sensory cell types. Functional analysis of TRPM homologs by RNA-interference (RNAi) revealed that disruption of Smed-TRPM-c expression results in reduced sperm development, indicating a role for this receptor in supporting spermatogenesis. Smed-TRPM-l RNAi did not result in a detectable phenotype, but it increased sperm development deficiencies when combined with Smed-TRPM-c RNAi. Fluorescence in situ hybridization revealed expression of Smed-TRPM-c in early spermatogenic cells within testes, suggesting cell-autonomous regulatory functions in germ cells for this gene. In addition, Smed-TRPM-c RNAi resulted in reduced numbers of presumptive germline stem cell clusters in asexual planarians, suggesting that Smed-TRPM-c supports establishment, maintenance, and/or expansion of spermatogonial germline stem cells. While further research is needed to identify the factors that trigger Smed-TRPM-c activity, these findings reveal one of few known examples for TRPM function in direct regulation of sperm development.
Collapse
Affiliation(s)
- Haley Nicole Curry
- Department of Biological Sciences, Wright State University, 3640 Colonel Glenn Hwy., Dayton, OH 45435, USA
| | - Roger Huynh
- Department of Biology, University of Massachusetts Boston, 100 William T. Morrissey Blvd., Boston, MA 02125-3393, USA
| | - Labib Rouhana
- Department of Biology, University of Massachusetts Boston, 100 William T. Morrissey Blvd., Boston, MA 02125-3393, USA
| |
Collapse
|
33
|
Sparta B, Hamilton T, Natesan G, Aragones SD, Deeds EJ. Binomial models uncover biological variation during feature selection of droplet-based single-cell RNA sequencing. PLoS Comput Biol 2024; 20:e1012386. [PMID: 39241106 PMCID: PMC11410258 DOI: 10.1371/journal.pcbi.1012386] [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: 01/30/2024] [Revised: 09/18/2024] [Accepted: 08/05/2024] [Indexed: 09/08/2024] Open
Abstract
Effective analysis of single-cell RNA sequencing (scRNA-seq) data requires a rigorous distinction between technical noise and biological variation. In this work, we propose a simple feature selection model, termed "Differentially Distributed Genes" or DDGs, where a binomial sampling process for each mRNA species produces a null model of technical variation. Using scRNA-seq data where cell identities have been established a priori, we find that the DDG model of biological variation outperforms existing methods. We demonstrate that DDGs distinguish a validated set of real biologically varying genes, minimize neighborhood distortion, and enable accurate partitioning of cells into their established cell-type groups.
Collapse
Affiliation(s)
- Breanne Sparta
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, United States of America
| | - Timothy Hamilton
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, United States of America
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, California, United States of America
| | - Gunalan Natesan
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, United States of America
| | - Samuel D Aragones
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, United States of America
| | - Eric J Deeds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, United States of America
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, California, United States of America
| |
Collapse
|
34
|
Maynard A, Soretić M, Treutlein B. Single-cell genomic profiling to study regeneration. Curr Opin Genet Dev 2024; 87:102231. [PMID: 39053027 DOI: 10.1016/j.gde.2024.102231] [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: 03/15/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/27/2024]
Abstract
Regenerative capacities and strategies vary dramatically across animals, as well as between cell types, organs, and with age. In recent years, high-throughput single-cell transcriptomics and other single-cell profiling technologies have been applied to many animal models to gain an understanding of the cellular and molecular mechanisms underlying regeneration. Here, we review recent single-cell studies of regeneration in diverse contexts and summarize key concepts that have emerged. The immense regenerative capacity of some invertebrates, exemplified by planarians, is driven mainly by the differentiation of abundant adult pluripotent stem cells, whereas in many other cases, regeneration involves the reactivation of embryonic or developmental gene-regulatory networks in differentiated cell types. However, regeneration also differs from development in many ways, including the use of regeneration-specific cell types and gene regulatory networks.
Collapse
Affiliation(s)
- Ashley Maynard
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Mateja Soretić
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland
| | - Barbara Treutlein
- ETH Zurich, Department of Biosystems Science and Engineering, Basel, Switzerland.
| |
Collapse
|
35
|
Lee JR, Boothe T, Mauksch C, Thommen A, Rink JC. Epidermal turnover in the planarian Schmidtea mediterranea involves basal cell extrusion and intestinal digestion. Cell Rep 2024; 43:114305. [PMID: 38906148 DOI: 10.1016/j.celrep.2024.114305] [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: 11/06/2023] [Revised: 03/21/2024] [Accepted: 05/15/2024] [Indexed: 06/23/2024] Open
Abstract
Planarian flatworms undergo continuous internal turnover, wherein old cells are replaced by the division progeny of adult pluripotent stem cells (neoblasts). How cell turnover is carried out at the organismal level remains an intriguing question in planarians and other systems. While previous studies have predominantly focused on neoblast proliferation, little is known about the processes that mediate cell loss during tissue homeostasis. Here, we use the planarian epidermis as a model to study the mechanisms of cell removal. We established a covalent dye-labeling assay and image analysis pipeline to quantify the cell turnover rate in the planarian epidermis. Our findings indicate that the ventral epidermis is highly dynamic and epidermal cells undergo internalization via basal extrusion, followed by a relocation toward the intestine and ultimately digestion by intestinal phagocytes. Overall, our study reveals a complex homeostatic process of cell clearance that may generally allow planarians to catabolize their own cells.
Collapse
Affiliation(s)
- Jun-Ru Lee
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany; Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, 37077 Göttingen, Germany
| | - Tobias Boothe
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Clemens Mauksch
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Albert Thommen
- Cancer Metabolism Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany; Faculty of Biology and Psychology, Georg-August-University, Göttingen, Germany.
| |
Collapse
|
36
|
Ross KG, Zepeda SA, Auwal MA, Garces AK, Roman S, Zayas RM. The role of polycystic kidney disease-like homologs in planarian nervous system regeneration and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603829. [PMID: 39091889 PMCID: PMC11291080 DOI: 10.1101/2024.07.17.603829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Planarians are an excellent model for investigating molecular mechanisms necessary for regenerating a functional nervous system. Numerous studies have led to the generation of extensive genomic resources, especially whole-animal single-cell RNA-seq resources. These have facilitated in silico predictions of neuronal subtypes, many of which have been anatomically mapped by in situ hybridization. However, our knowledge of the function of dozens of neuronal subtypes remains poorly understood. Previous investigations identified that polycystic kidney disease (pkd)-like genes in planarians are strongly expressed in sensory neurons and have roles in mechanosensation. Here, we examine the expression and function of all the pkd genes found in the Schmidtea mediterranea genome and map their expression in the asexual and hermaphroditic strains. Using custom behavioral assays, we test the function of pkd genes in response to mechanical stimulation and in food detection. Our work provides insight into the physiological function of sensory neuron populations and protocols for creating inexpensive automated setups for acquiring and analyzing mechanosensory stimulation in planarians.
Collapse
Affiliation(s)
- Kelly G. Ross
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Sarai Alvarez Zepeda
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Mohammad A. Auwal
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Audrey K. Garces
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Sydney Roman
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| | - Ricardo M. Zayas
- Department of Biology, San Diego State University, 5500 Campanile Dr., San Diego, CA 92182-4614, USA
| |
Collapse
|
37
|
Grobecker P, Sakoparnig T, van Nimwegen E. Identifying cell states in single-cell RNA-seq data at statistically maximal resolution. PLoS Comput Biol 2024; 20:e1012224. [PMID: 38995959 PMCID: PMC11364423 DOI: 10.1371/journal.pcbi.1012224] [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: 11/06/2023] [Revised: 08/30/2024] [Accepted: 06/04/2024] [Indexed: 07/14/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has become a popular experimental method to study variation of gene expression within a population of cells. However, obtaining an accurate picture of the diversity of distinct gene expression states that are present in a given dataset is highly challenging because of the sparsity of the scRNA-seq data and its inhomogeneous measurement noise properties. Although a vast number of different methods is applied in the literature for clustering cells into subsets with 'similar' expression profiles, these methods generally lack rigorously specified objectives, involve multiple complex layers of normalization, filtering, feature selection, dimensionality-reduction, employ ad hoc measures of distance or similarity between cells, often ignore the known measurement noise properties of scRNA-seq measurements, and include a large number of tunable parameters. Consequently, it is virtually impossible to assign concrete biophysical meaning to the clusterings that result from these methods. Here we address the following problem: Given raw unique molecule identifier (UMI) counts of an scRNA-seq dataset, partition the cells into subsets such that the gene expression states of the cells in each subset are statistically indistinguishable, and each subset corresponds to a distinct gene expression state. That is, we aim to partition cells so as to maximally reduce the complexity of the dataset without removing any of its meaningful structure. We show that, given the known measurement noise structure of scRNA-seq data, this problem is mathematically well-defined and derive its unique solution from first principles. We have implemented this solution in a tool called Cellstates which operates directly on the raw data and automatically determines the optimal partition and cluster number, with zero tunable parameters. We show that, on synthetic datasets, Cellstates almost perfectly recovers optimal partitions. On real data, Cellstates robustly identifies subtle substructure within groups of cells that are traditionally annotated as a common cell type. Moreover, we show that the diversity of gene expression states that Cellstates identifies systematically depends on the tissue of origin and not on technical features of the experiments such as the total number of cells and total UMI count per cell. In addition to the Cellstates tool we also provide a small toolbox of software to place the identified cellstates into a hierarchical tree of higher-order clusters, to identify the most important differentially expressed genes at each branch of this hierarchy, and to visualize these results.
Collapse
Affiliation(s)
- Pascal Grobecker
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Thomas Sakoparnig
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Erik van Nimwegen
- Biozentrum, University of Basel and Swiss Institute of Bioinformatics, Basel, Switzerland
| |
Collapse
|
38
|
Issigonis M, Browder KL, Chen R, Collins JJ, Newmark PA. A niche-derived nonribosomal peptide triggers planarian sexual development. Proc Natl Acad Sci U S A 2024; 121:e2321349121. [PMID: 38889152 PMCID: PMC11214079 DOI: 10.1073/pnas.2321349121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
Germ cells are regulated by local microenvironments (niches), which secrete instructive cues. Conserved developmental signaling molecules act as niche-derived regulatory factors, yet other types of niche signals remain to be identified. Single-cell RNA-sequencing of sexual planarians revealed niche cells expressing a nonribosomal peptide synthetase (nrps). Inhibiting nrps led to loss of female reproductive organs and testis hyperplasia. Mass spectrometry detected the dipeptide β-alanyl-tryptamine (BATT), which is associated with reproductive system development and requires nrps and a monoamine-transmitter-synthetic enzyme Aromatic L-amino acid decarboxylase (AADC) for its production. Exogenous BATT rescued the reproductive defects after nrps or aadc inhibition, restoring fertility. Thus, a nonribosomal, monoamine-derived peptide provided by niche cells acts as a critical signal to trigger planarian reproductive development. These findings reveal an unexpected function for monoamines in niche-germ cell signaling. Furthermore, given the recently reported role for BATT as a male-derived factor required for reproductive maturation of female schistosomes, these results have important implications for the evolution of parasitic flatworms and suggest a potential role for nonribosomal peptides as signaling molecules in other organisms.
Collapse
Affiliation(s)
- Melanie Issigonis
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI53715
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI53715
| | - Katherine L. Browder
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI53715
- HMI, University of Wisconsin-Madison, Madison, WI53715
| | - Rui Chen
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - James J. Collins
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Phillip A. Newmark
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI53715
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI53715
- HMI, University of Wisconsin-Madison, Madison, WI53715
| |
Collapse
|
39
|
Gąsiorowski L, Chai C, Rozanski A, Purandare G, Ficze F, Mizi A, Wang B, Rink JC. Regeneration in the absence of canonical neoblasts in an early branching flatworm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595708. [PMID: 38853907 PMCID: PMC11160568 DOI: 10.1101/2024.05.24.595708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
The remarkable regenerative abilities of flatworms are closely linked to neoblasts - adult pluripotent stem cells that are the only division-competent cell type outside of the reproductive system. Although the presence of neoblast-like cells and whole-body regeneration in other animals has led to the idea that these features may represent the ancestral metazoan state, the evolutionary origin of both remains unclear. Here we show that the catenulid Stenostomum brevipharyngium, a member of the earliest-branching flatworm lineage, lacks conventional neoblasts despite being capable of whole-body regeneration and asexual reproduction. Using a combination of single-nuclei transcriptomics, in situ gene expression analysis, and functional experiments, we find that cell divisions are not restricted to a single cell type and are associated with multiple fully differentiated somatic tissues. Furthermore, the cohort of germline multipotency genes, which are considered canonical neoblast markers, are not expressed in dividing cells, but in the germline instead, and we experimentally show that they are neither necessary for proliferation nor regeneration. Overall, our results challenge the notion that canonical neoblasts are necessary for flatworm regeneration and open up the possibility that neoblast-like cells may have evolved convergently in different animals, independent of their regenerative capacity.
Collapse
Affiliation(s)
- Ludwik Gąsiorowski
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Chew Chai
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Andrei Rozanski
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gargi Purandare
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Fruzsina Ficze
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Athanasia Mizi
- Institute of Pathology, University Medical Centre Göttingen, Göttingen, Germany
| | - Bo Wang
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Jochen C Rink
- Department of Tissue Dynamics and Regeneration, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
40
|
Verma P, Sánchez Alvarado A, Duncan EM. Chromatin remodeling protein BPTF regulates transcriptional stability in planarian stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595819. [PMID: 38826365 PMCID: PMC11142235 DOI: 10.1101/2024.05.24.595819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Trimethylation of histone H3 lysine 4 (H3K4me3) correlates strongly with gene expression in many different organisms, yet the question of whether it plays a causal role in transcriptional activity remains unresolved. Although H3K4me3 does not directly affect chromatin accessibility, it can indirectly affect genome accessibility by recruiting the ATP-dependent chromatin remodeling complex NuRF (Nucleosome Remodeling Factor). The largest subunit of NuRF, BPTF/NURF301, binds H3K4me3 specifically and recruits the NuRF complex to loci marked by this modification. Studies have shown that the strength and duration of BPTF binding likely also depends on additional chromatin features at these loci, such as lysine acetylation and variant histone proteins. However, the exact details of this recruitment mechanism vary between studies and have largely been tested in vitro. Here, we use stem cells isolated directly from live planarian animals to investigate the role of BPTF in regulating chromatin accessibility in vivo. We find that BPTF operates at gene promoters and is most effective at facilitating transcription at genes marked by Set1-dependent H3K4me3 peaks, which are significantly broader than those added by the lysine methyltransferase MLL1/2. Moreover, BPTF is essential for planarian stem cell biology and its loss of function phenotype mimics that of Set1 knockdown. Together, these data suggest that BPTF and H3K4me3 are important mediators of both transcription and in vivo stem cell function.
Collapse
|
41
|
Doddihal V, Mann FG, Ross EJ, McKinney MC, Guerrero-Hernández C, Brewster CE, McKinney SA, Sánchez Alvarado A. A PAK family kinase and the Hippo/Yorkie pathway modulate WNT signaling to functionally integrate body axes during regeneration. Proc Natl Acad Sci U S A 2024; 121:e2321919121. [PMID: 38713625 PMCID: PMC11098123 DOI: 10.1073/pnas.2321919121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/03/2024] [Indexed: 05/09/2024] Open
Abstract
Successful regeneration of missing tissues requires seamless integration of positional information along the body axes. Planarians, which regenerate from almost any injury, use conserved, developmentally important signaling pathways to pattern the body axes. However, the molecular mechanisms which facilitate cross talk between these signaling pathways to integrate positional information remain poorly understood. Here, we report a p21-activated kinase (smed-pak1) which functionally integrates the anterior-posterior (AP) and the medio-lateral (ML) axes. pak1 inhibits WNT/β-catenin signaling along the AP axis and, functions synergistically with the β-catenin-independent WNT signaling of the ML axis. Furthermore, this functional integration is dependent on warts and merlin-the components of the Hippo/Yorkie (YKI) pathway. Hippo/YKI pathway is a critical regulator of body size in flies and mice, but our data suggest the pathway regulates body axes patterning in planarians. Our study provides a signaling network integrating positional information which can mediate coordinated growth and patterning during planarian regeneration.
Collapse
Affiliation(s)
- Viraj Doddihal
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | - Eric J. Ross
- Stowers Institute for Medical Research, Kansas City, MO64110
| | | | | | | | | | | |
Collapse
|
42
|
Morizet D, Foucher I, Alunni A, Bally-Cuif L. Reconstruction of macroglia and adult neurogenesis evolution through cross-species single-cell transcriptomic analyses. Nat Commun 2024; 15:3306. [PMID: 38632253 PMCID: PMC11024210 DOI: 10.1038/s41467-024-47484-1] [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: 04/14/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024] Open
Abstract
Macroglia fulfill essential functions in the adult vertebrate brain, producing and maintaining neurons and regulating neuronal communication. However, we still know little about their emergence and diversification. We used the zebrafish D. rerio as a distant vertebrate model with moderate glial diversity as anchor to reanalyze datasets covering over 600 million years of evolution. We identify core features of adult neurogenesis and innovations in the mammalian lineage with a potential link to the rarity of radial glia-like cells in adult humans. Our results also suggest that functions associated with astrocytes originated in a multifunctional cell type fulfilling both neural stem cell and astrocytic functions before these diverged. Finally, we identify conserved elements of macroglial cell identity and function and their time of emergence during evolution.
Collapse
Affiliation(s)
- David Morizet
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France.
- Sorbonne Université, Collège doctoral, F-75005, Paris, France.
| | - Isabelle Foucher
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France
| | - Alessandro Alunni
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France
- Institut des Neurosciences Paris-Saclay, Université Paris-Saclay, CNRS UMR9197, F-91190, Gif-sur-Yvette, France
| | - Laure Bally-Cuif
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Zebrafish Neurogenetics Unit, Team supported by the Ligue Nationale Contre le Cancer, F-75015, Paris, France.
| |
Collapse
|
43
|
Álvarez-Campos P, García-Castro H, Emili E, Pérez-Posada A, Del Olmo I, Peron S, Salamanca-Díaz DA, Mason V, Metzger B, Bely AE, Kenny NJ, Özpolat BD, Solana J. Annelid adult cell type diversity and their pluripotent cellular origins. Nat Commun 2024; 15:3194. [PMID: 38609365 PMCID: PMC11014941 DOI: 10.1038/s41467-024-47401-6] [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: 07/28/2023] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Many annelids can regenerate missing body parts or reproduce asexually, generating all cell types in adult stages. However, the putative adult stem cell populations involved in these processes, and the diversity of cell types generated by them, are still unknown. To address this, we recover 75,218 single cell transcriptomes of the highly regenerative and asexually-reproducing annelid Pristina leidyi. Our results uncover a rich cell type diversity including annelid specific types as well as novel types. Moreover, we characterise transcription factors and gene networks that are expressed specifically in these populations. Finally, we uncover a broadly abundant cluster of putative stem cells with a pluripotent signature. This population expresses well-known stem cell markers such as vasa, piwi and nanos homologues, but also shows heterogeneous expression of differentiated cell markers and their transcription factors. We find conserved expression of pluripotency regulators, including multiple chromatin remodelling and epigenetic factors, in piwi+ cells. Finally, lineage reconstruction analyses reveal computational differentiation trajectories from piwi+ cells to diverse adult types. Our data reveal the cell type diversity of adult annelids by single cell transcriptomics and suggest that a piwi+ cell population with a pluripotent stem cell signature is associated with adult cell type differentiation.
Collapse
Affiliation(s)
- Patricia Álvarez-Campos
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK.
- Centro de Investigación en Biodiversidad y Cambio Global (CIBC-UAM) & Departamento de Biología (Zoología), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Helena García-Castro
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
| | - Elena Emili
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Alberto Pérez-Posada
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
| | - Irene Del Olmo
- Centro de Investigación en Biodiversidad y Cambio Global (CIBC-UAM) & Departamento de Biología (Zoología), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sophie Peron
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
| | - David A Salamanca-Díaz
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Living Systems Institute, University of Exeter, Exeter, UK
| | - Vincent Mason
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Bria Metzger
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA, 05432, USA
- Department of Biology, Washington University in St. Louis. 1 Brookings Dr. Saint Louis, Saint Louis, MO, 63130, USA
| | - Alexandra E Bely
- Department of Biology, University of Maryland, College Park, MD, 20742, USA
| | - Nathan J Kenny
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
- Department of Biochemistry, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand
| | - B Duygu Özpolat
- Eugene Bell Center for Regenerative Biology and Tissue Engineering, Marine Biological Laboratory, 7 MBL Street, Woods Hole, MA, 05432, USA.
- Department of Biology, Washington University in St. Louis. 1 Brookings Dr. Saint Louis, Saint Louis, MO, 63130, USA.
| | - Jordi Solana
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK.
- Living Systems Institute, University of Exeter, Exeter, UK.
| |
Collapse
|
44
|
Tyagi R, Rosa BA, Swain A, Artyomov MN, Jasmer DP, Mitreva M. Intestinal cell diversity and treatment responses in a parasitic nematode at single cell resolution. BMC Genomics 2024; 25:341. [PMID: 38575858 PMCID: PMC10996262 DOI: 10.1186/s12864-024-10203-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/08/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Parasitic nematodes, significant pathogens for humans, animals, and plants, depend on diverse organ systems for intra-host survival. Understanding the cellular diversity and molecular variations underlying these functions holds promise for developing novel therapeutics, with specific emphasis on the neuromuscular system's functional diversity. The nematode intestine, crucial for anthelmintic therapies, exhibits diverse cellular phenotypes, and unraveling this diversity at the single-cell level is essential for advancing knowledge in anthelmintic research across various organ systems. RESULTS Here, using novel single-cell transcriptomics datasets, we delineate cellular diversity within the intestine of adult female Ascaris suum, a parasitic nematode species that infects animals and people. Gene transcripts expressed in individual nuclei of untreated intestinal cells resolved three phenotypic clusters, while lower stringency resolved additional subclusters and more potential diversity. Clusters 1 and 3 phenotypes displayed variable congruence with scRNA phenotypes of C. elegans intestinal cells, whereas the A. suum cluster 2 phenotype was markedly unique. Distinct functional pathway enrichment characterized each A. suum intestinal cell cluster. Cluster 2 was distinctly enriched for Clade III-associated genes, suggesting it evolved within clade III nematodes. Clusters also demonstrated differential transcriptional responsiveness to nematode intestinal toxic treatments, with Cluster 2 displaying the least responses to short-term intra-pseudocoelomic nematode intestinal toxin treatments. CONCLUSIONS This investigation presents advances in knowledge related to biological differences among major cell populations of adult A. suum intestinal cells. For the first time, diverse nematode intestinal cell populations were characterized, and associated biological markers of these cells were identified to support tracking of constituent cells under experimental conditions. These advances will promote better understanding of this and other parasitic nematodes of global importance, and will help to guide future anthelmintic treatments.
Collapse
Affiliation(s)
- Rahul Tyagi
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, 63110, St. Louis, MO, USA
| | - Bruce A Rosa
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, 63110, St. Louis, MO, USA
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, 63110, Saint Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, 63110, Saint Louis, MO, USA
| | - Douglas P Jasmer
- Department of Veterinary Microbiology and Pathology, Washington State University, 99164, Pullman, WA, USA.
| | - Makedonka Mitreva
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, 63110, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, 63110, St. Louis, MO, USA.
- McDonnell Genome Institute, Washington University School of Medicine, 63110, St Louis, MO, USA.
| |
Collapse
|
45
|
King HO, Owusu-Boaitey KE, Fincher CT, Reddien PW. A transcription factor atlas of stem cell fate in planarians. Cell Rep 2024; 43:113843. [PMID: 38401119 PMCID: PMC11232438 DOI: 10.1016/j.celrep.2024.113843] [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: 04/05/2023] [Revised: 12/05/2023] [Accepted: 02/06/2024] [Indexed: 02/26/2024] Open
Abstract
Whole-body regeneration requires the ability to produce the full repertoire of adult cell types. The planarian Schmidtea mediterranea contains over 125 cell types, which can be regenerated from a stem cell population called neoblasts. Neoblast fate choice can be regulated by the expression of fate-specific transcription factors (FSTFs). How fate choices are made and distributed across neoblasts versus their post-mitotic progeny remains unclear. We used single-cell RNA sequencing to systematically map fate choices made in S/G2/M neoblasts and, separately, in their post-mitotic progeny that serve as progenitors for all adult cell types. We defined transcription factor expression signatures associated with all detected fates, identifying numerous new progenitor classes and FSTFs that regulate them. Our work generates an atlas of stem cell fates with associated transcription factor signatures for most cell types in a complete adult organism.
Collapse
Affiliation(s)
- Hunter O King
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kwadwo E Owusu-Boaitey
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA; Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Christopher T Fincher
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Peter W Reddien
- Howard Hughes Medical Institute, Chevy Chase, MD, USA; Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
46
|
Allikka Parambil S, Li D, Zelko M, Poulet A, van Wolfswinkel J. piRNA generation is associated with the pioneer round of translation in stem cells. Nucleic Acids Res 2024; 52:2590-2608. [PMID: 38142432 PMCID: PMC10954484 DOI: 10.1093/nar/gkad1212] [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: 04/13/2023] [Revised: 12/05/2023] [Accepted: 12/08/2023] [Indexed: 12/26/2023] Open
Abstract
Much insight has been gained on how stem cells maintain genomic integrity, but less attention has been paid to how they maintain their transcriptome. Here, we report that the PIWI protein SMEDWI-1 plays a role in the filtering of dysfunctional transcripts from the transcriptome of planarian stem cells. SMEDWI-1 accomplishes this through association with the ribosomes during the pioneer round of translation, and processing of poorly translated transcripts into piRNAs. This results in the removal of such transcripts from the cytoplasmic pool and at the same time creates a dynamic pool of small RNAs for post-transcriptional surveillance through the piRNA pathway. Loss of SMEDWI-1 results in elevated levels of several non-coding transcripts, including rRNAs, snRNAs and pseudogene mRNAs, while reducing levels of several coding transcripts. In the absence of SMEDWI-1, stem cell colonies are delayed in their expansion and a higher fraction of descendants exit the stem cell state, indicating that this transcriptomic sanitation mediated by SMEDWI-1 is essential to maintain stem cell health. This study presents a new model for the function of PIWI proteins in stem cell maintenance, that complements their role in transposon repression, and proposes a new biogenesis pathway for piRNAs in stem cells.
Collapse
Affiliation(s)
- Sudheesh Allikka Parambil
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06511, USA
- Center for RNA science and medicine, Yale School of Medicine, New Haven. CT 06511, USA
| | - Danyan Li
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06511, USA
- Center for RNA science and medicine, Yale School of Medicine, New Haven. CT 06511, USA
| | - Michael Zelko
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06511, USA
- Center for RNA science and medicine, Yale School of Medicine, New Haven. CT 06511, USA
| | - Axel Poulet
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06511, USA
- Center for RNA science and medicine, Yale School of Medicine, New Haven. CT 06511, USA
| | - Josien C van Wolfswinkel
- Department of Molecular Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06511, USA
- Center for RNA science and medicine, Yale School of Medicine, New Haven. CT 06511, USA
| |
Collapse
|
47
|
Costábile A, Domínguez MF, Guarnaschelli I, Preza M, Koziol U, Castillo E, Tort JF. Purification and transcriptomic characterization of proliferative cells of Mesocestoides corti selectively affected by irradiation. FRONTIERS IN PARASITOLOGY 2024; 3:1362199. [PMID: 39817174 PMCID: PMC11732142 DOI: 10.3389/fpara.2024.1362199] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/12/2024] [Indexed: 01/18/2025]
Abstract
Flatworms depend on stem cells for continued tissue growth and renewal during their life cycles, making these cells valuable drug targets. While neoblasts are extensively characterized in the free-living planarian Schmidtea mediterranea, and similar stem cells have been characterized in the trematode Schistosoma mansoni, their identification and characterization in cestodes is just emerging. Since stem cells are generally affected by irradiation, in this work we used this experimental approach to study the stem cells of the model cestode Mesocestoides corti. We found that gamma irradiation produces a dose-dependent decrease in proliferative cells, requiring higher doses than in other flatworms to completely abolish proliferation. The treatment results in the downregulation of candidate marker genes. Transcriptomic studies reveal that several genes downregulated after irradiation are conserved with other flatworms, and are related to cell cycle, DNA replication and repair functions. Furthermore, proliferative cells were isolated by cell sorting and also characterized transcriptomically. We found that the set of genes characteristic of proliferative cells agrees well with those downregulated during irradiation, and have a significant overlap with those expressed in planarian neoblasts or S. mansoni stem cells. Our study highlights that conserved mechanisms of stem cell biology may be functional in flatworms, suggesting that these could be relevant targets to evaluate in the control of parasitic species.
Collapse
Affiliation(s)
- Alicia Costábile
- Sección Bioquímica, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - María Fernanda Domínguez
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Inés Guarnaschelli
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Matías Preza
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Uriel Koziol
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Estela Castillo
- Unidad de Biología Parasitaria, Facultad de Ciencias- Instituto de Higiene, Universidad de la República, Montevideo, Uruguay
| | - José F. Tort
- Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
48
|
Wilson MM, Roberts-Galbraith RH. In preprints: allometry of cell types during animal growth and degrowth. Development 2024; 151:dev202790. [PMID: 38415753 PMCID: PMC10941661 DOI: 10.1242/dev.202790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Affiliation(s)
- Macey M. Wilson
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | | |
Collapse
|
49
|
Ye F, Wang J, Li J, Mei Y, Guo G. Mapping Cell Atlases at the Single-Cell Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305449. [PMID: 38145338 PMCID: PMC10885669 DOI: 10.1002/advs.202305449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/01/2023] [Indexed: 12/26/2023]
Abstract
Recent advancements in single-cell technologies have led to rapid developments in the construction of cell atlases. These atlases have the potential to provide detailed information about every cell type in different organisms, enabling the characterization of cellular diversity at the single-cell level. Global efforts in developing comprehensive cell atlases have profound implications for both basic research and clinical applications. This review provides a broad overview of the cellular diversity and dynamics across various biological systems. In addition, the incorporation of machine learning techniques into cell atlas analyses opens up exciting prospects for the field of integrative biology.
Collapse
Affiliation(s)
- Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jiaqi Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Yuqing Mei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative MedicineDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineHangzhouZhejiang310058China
- Institute of HematologyZhejiang UniversityHangzhouZhejiang310000China
| |
Collapse
|
50
|
Herz M, Zarowiecki M, Wessels L, Pätzel K, Herrmann R, Braun C, Holroyd N, Huckvale T, Bergmann M, Spiliotis M, Koziol U, Berriman M, Brehm K. Genome-wide transcriptome analysis of Echinococcus multilocularis larvae and germinative cell cultures reveals genes involved in parasite stem cell function. Front Cell Infect Microbiol 2024; 14:1335946. [PMID: 38333034 PMCID: PMC10850878 DOI: 10.3389/fcimb.2024.1335946] [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: 11/09/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
The lethal zoonosis alveolar echinococcosis is caused by tumour-like growth of the metacestode stage of the tapeworm Echinococcus multilocularis within host organs. We previously demonstrated that metacestode proliferation is exclusively driven by somatic stem cells (germinative cells), which are the only mitotically active parasite cells that give rise to all differentiated cell types. The Echinococcus gene repertoire required for germinative cell maintenance and differentiation has not been characterised so far. We herein carried out Illumina sequencing on cDNA from Echinococcus metacestode vesicles, from metacestode tissue depleted of germinative cells, and from Echinococcus primary cell cultures. We identified a set of ~1,180 genes associated with germinative cells, which contained numerous known stem cell markers alongside genes involved in replication, cell cycle regulation, mitosis, meiosis, epigenetic modification, and nucleotide metabolism. Interestingly, we also identified 44 stem cell associated transcription factors that are likely involved in regulating germinative cell differentiation and/or pluripotency. By in situ hybridization and pulse-chase experiments, we also found a new general Echinococcus stem cell marker, EmCIP2Ah, and we provide evidence implying the presence of a slow cycling stem cell sub-population expressing the extracellular matrix factor Emkal1. RNA-Seq analyses on primary cell cultures revealed that metacestode-derived Echinococcus stem cells display an expanded differentiation capability and do not only form differentiated cell types of the metacestode, but also cells expressing genes specific for protoscoleces, adult worms, and oncospheres, including an ortholog of the schistosome praziquantel target, EmTRPMPZQ. Finally, we show that primary cell cultures contain a cell population expressing an ortholog of the tumour necrosis factor α receptor family and that mammalian TNFα accelerates the development of metacestode vesicles from germinative cells. Taken together, our analyses provide a robust and comprehensive characterization of the Echinococcus germinative cell transcriptome, demonstrate expanded differentiation capability of metacestode derived stem cells, and underscore the potential of primary germinative cell cultures to investigate developmental processes of the parasite. These data are relevant for studies into the role of Echinococcus stem cells in parasite development and will facilitate the design of anti-parasitic drugs that specifically act on the parasite germinative cell compartment.
Collapse
Affiliation(s)
- Michaela Herz
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | | | - Leonie Wessels
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Katharina Pätzel
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Ruth Herrmann
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Christiane Braun
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Nancy Holroyd
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Thomas Huckvale
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Monika Bergmann
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Markus Spiliotis
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Uriel Koziol
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
- Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Matthew Berriman
- Parasite Genomics, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Klaus Brehm
- Consultant Laboratory for Echinococcosis, Institute of Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|