1
|
LEE WK, WAN HT, CHENG Z, CHAN WY, LAM TKY, LAI KP, WANG J, CAI Z, WONG CKC. Impact of PFOS Exposure on Murine Fetal Hematopoietic Stem Cells, Associated with Intrauterine Metabolic Perturbation. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:5496-5509. [PMID: 40082253 PMCID: PMC11948485 DOI: 10.1021/acs.est.5c02623] [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: 02/25/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025]
Abstract
This study hypothesized that perfluorooctanesulfonate (PFOS) exposure disrupts maternal-fetal metabolism, affecting fetal liver hematopoietic stem cell (FL-HSC) development. Pregnant mice received PFOS (0.3 and 3 μg/g bw) and were sacrificed on gestation day 14.5. Metabolomic analysis of maternal plasma revealed disruptions in steroid hormone, purine, carbohydrate, and amino acid metabolism, which aligned with the enriched pathways in amniotic fluid (AF). FL analysis indicated increased purine metabolism and disrupted glucose and amino acid metabolism. FL exhibited higher levels of polyunsaturated fatty acids, glycolytic and TCA metabolites, and pro-inflammatory cytokine IL-23, crucial for hematopoiesis regulation. Transcriptomic analysis of FL-HSCs revealed disturbances in the PPAR signaling pathway, pyruvate metabolism, oxidative phosphorylation, and amino acid metabolism, correlating with FL metabolic changes. Metabolomic analysis indicated significant rises in glycerophospholipid and vitamin B6 metabolism related to HSC expansion and differentiation. Flow cytometric analysis confirmed increased HSC populations and progenitor activation for megakaryocyte, erythrocyte, and lymphocyte lineages. The CFU assay showed a significant increase in BFU-E and CFU-G, but a decrease in CFU-GM in FL-HSCs from the H-PFOS group, indicating altered differentiation potential. These findings provide for the first time insights into the effects of PFOS on maternal-fetal metabolism and fetal hematopoiesis, highlighting implications for pollution-affected immune functions.
Collapse
Affiliation(s)
- Wang Ka LEE
- Croucher
Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR
| | - Hin Ting WAN
- Croucher
Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR
| | - Zheyu CHENG
- Croucher
Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR
| | - Wing Yee CHAN
- Croucher
Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR
| | - Thomas Ka Yam LAM
- State
Key Laboratory in Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR
| | - Keng Po LAI
- Department
of Applied Science, Hong Kong Metropolitan
University, Hong Kong SAR
| | - Jianing WANG
- State
Key Laboratory in Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR
| | - Zongwei CAI
- State
Key Laboratory in Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR
| | - Chris Kong Chu WONG
- Croucher
Institute for Environmental Sciences, Department of Biology, Hong Kong Baptist University, Hong Kong SAR
- State
Key Laboratory in Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong SAR
| |
Collapse
|
2
|
Wang D, Shu W. Protein expression changes in Tibetan middle-to-long distance runners after the transition from high altitude to low altitude: Implications for enhancing endurance training. SPORTS MEDICINE AND HEALTH SCIENCE 2024; 6:370-377. [PMID: 39309459 PMCID: PMC11411288 DOI: 10.1016/j.smhs.2023.12.005] [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: 01/15/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 09/25/2024] Open
Abstract
The study aims to investigate the differences in protein expressions in Xizang's (Tibetan) middle-to-long distance runners after the transition from high altitude to low altitude and reveal the molecular mechanisms underlying their enhanced middle-to-long distance running performance. In the study, eleven subjects were selected from native Tibetan middle-to-long distance runners to participate in an 8-week pre-competition exercise training program consisting of a 6-week training stage in Kangding City at an altitude of 2 560 meters (m) and a subsequent 2-week training stage in Leshan City at an altitude of 360 m. Blood samples were collected twice from the runners before beginning altitude exercise training in Kangding and after going to sea level - Leshan City. Using a label-free quantitative method, peptides in the samples were analyzed by mass spectrometry. Proteomic analysis was performed to identify differentially expressed proteins and predict their biological functions. A total of 846 proteins were identified in the 21 samples, including 719 quantified proteins. In total, 49 significantly differentially expressed proteins (p < 0.05) were identified, including twenty-eight 0.2-fold up-regulated proteins or twenty-one 0.17-fold down-regulated proteins. The up-regulated proteins, including cystic fibrosis transmembrane conductance regulator (CFTR) and carbonic anhydrase I (CAI), were of particular interest due to their role in regulating the oxygen saturation in deep tissues. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that these proteins were mainly involved in regulating actin cytoskeleton, local adhesion, biotin absorption and metabolism, immune system, cancer, and membrane transport processes. In conclusion, Tibetan middle-to-long distance runners who resided in high-altitude areas benefited from repeated plateau-plain alternate training mode during the pre-competition period. The training mode induced positive changes in peripheral blood plasma proteins (CFTR and CAI), the biomarkers associated with aerobic capacity. Among the 11 runners, one female athlete won the gold medal in the 3 000-m running event in this competition, demonstrating that the plateau-plain alternate training mode could enhance the aerobic capacity of athletes.
Collapse
Affiliation(s)
- Di Wang
- Chengdu Sport University, Chengdu, Sichuan, China
| | - Weiping Shu
- Chengdu Sport University, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Xiu Y, Xiong M, Yang H, Wang Q, Zhao X, Long J, Liang F, Liu N, Chen F, Gao M, Sun Y, Fan R, Zeng Y. Proteomic characterization of murine hematopoietic stem progenitor cells reveals dynamic fetal-to-adult changes in metabolic-related pathways. Biochem Biophys Res Commun 2024; 734:150661. [PMID: 39243675 DOI: 10.1016/j.bbrc.2024.150661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/24/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Hematopoietic stem progenitor cells (HSPCs) give rise to the hematopoietic system, maintain hematopoiesis throughout the lifespan, and undergo molecular and functional changes during their development and aging. The importance of hematopoietic stem cell (HSC) biology has led to their extensive characterization at genomic and transcriptomic levels. However, the proteomics of HSPCs throughout the murine lifetime still needs to be fully completed. Here, using mass spectrometry (MS)-based quantitative proteomics, we report on the dynamic changes in the proteome of HSPCs from four developmental stages in the fetal liver (FL) and the bone marrow (BM), including E14.5, young (2 months), middle-aged (8 months), and aging (18 months) stages. Proteomics unveils highly dynamic protein kinetics during the development and aging of HSPCs. Our data identify stage-specific developmental features of HSPCs, which can be linked to their functional maturation and senescence. Our proteomic data demonstrated that FL HSPCs depend on aerobic respiration to meet their proliferation and oxygen supply demand, while adult HSPCs prefer glycolysis to preserve the HSC pool. By functional assays, we validated the decreased mitochondrial metabolism, glucose uptake, reactive oxygen species (ROS) production, protein synthesis rate, and increased glutathione S-transferase (GST) activity during HSPC development from fetal to adult. Distinct metabolism pathways and immune-related pathways enriched in different HSPC developmental stages were revealed at the protein level. Our study will have broader implications for understanding the mechanism of stem cell maintenance and fate determination and reversing the HSC aging process.
Collapse
Affiliation(s)
- Yanyu Xiu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Mingfang Xiong
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Haoyu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China; Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Qianqian Wang
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Xiao Zhao
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Juan Long
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Fei Liang
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Nan Liu
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Fudong Chen
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Meng Gao
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Yuying Sun
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, 030801, China.
| | - Yang Zeng
- Senior Department of Hematology, the Fifth Medical Center of PLA General Hospital, Beijing, 100071, China; Medical School of the Chinese PLA General Hospital, Beijing, 100039, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
4
|
Zhang Q, Li MK, Hu XY, Wu YX, Wang YY, Zhao PP, Cheng LN, Yu RH, Zhang XD, Chen S, Zhu ZM, de Bock CE, Thorne RF. The tumor suppressor Fat1 is dispensable for normal murine hematopoiesis. J Leukoc Biol 2024; 116:909-914. [PMID: 38833591 DOI: 10.1093/jleuko/qiae128] [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: 11/13/2023] [Revised: 04/29/2024] [Accepted: 05/16/2024] [Indexed: 06/06/2024] Open
Abstract
Loss and overexpression of FAT1 occurs among different cancers, with these divergent states equated with tumor suppressor and oncogene activity, respectively. Regarding the latter, FAT1 is highly expressed in a high proportion of human acute leukemias relative to normal blood cells, with evidence pointing to an oncogenic role. We hypothesized that this occurrence represents legacy expression of FAT1 in undefined hematopoietic precursor subsets (i.e. sustained following transformation), predicating a role for FAT1 during normal hematopoiesis. We explored this concept by using the Vav-iCre strain to construct conditional knockout mice in which Fat1 expression was deleted at the hematopoietic stem cell stage. Extensive analysis of precursor and mature blood populations using multipanel flow cytometry revealed no ostensible differences between Fat1 conditional knockout mice and normal littermates. Further functional comparisons involving colony-forming unit and competitive bone marrow transplantation assays support the conclusion that Fat1 is dispensable for normal murine hematopoiesis.
Collapse
Affiliation(s)
- Qing Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Meng Ke Li
- Institute of Hematology, Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Xin Yuan Hu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Yu Xin Wu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Ying Ying Wang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Pan Pan Zhao
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Lin Na Cheng
- Institute of Hematology, Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Rong Hua Yu
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Xu Dong Zhang
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Song Chen
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Zun Min Zhu
- Institute of Hematology, Henan Key Laboratory of Stem Cell Clinical Application and Key Technology, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| | - Charles E de Bock
- Children's Cancer Institute, Lowy Cancer Research Centre, UNSW Sydney, Cnr Botany & High Sts, Kensington, NSW 2031, Australia
- School of Clinical Medicine, UNSW Sydney, High St Kensington, NSW 2052, Australia
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Wei Wu Road, Jinshui District, Zhengzhou 450003, China
| |
Collapse
|
5
|
Sashittal P, Zhang RY, Law BK, Strzalkowski A, Schmidt H, Bolondi A, Chan MM, Raphael BJ. Inferring cell differentiation maps from lineage tracing data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.611835. [PMID: 39314473 PMCID: PMC11419031 DOI: 10.1101/2024.09.09.611835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
During development, mulitpotent cells differentiate through a hierarchy of increasingly restricted progenitor cell types until they realize specialized cell types. A cell differentiation map describes this hierarchy, and inferring these maps is an active area of research spanning traditional single marker lineage studies to data-driven trajectory inference methods on single-cell RNA-seq data. Recent high-throughput lineage tracing technologies profile lineages and cell types at scale, but current methods to infer cell differentiation maps from these data rely on simple models with restrictive assumptions about the developmental process. We introduce a mathematical framework for cell differentiation maps based on the concept of potency, and develop an algorithm, Carta, that infers an optimal cell differentiation map from single-cell lineage tracing data. The key insight in Carta is to balance the trade-off between the complexity of the cell differentiation map and the number of unobserved cell type transitions on the lineage tree. We show that Carta more accurately infers cell differentiation maps on both simulated and real data compared to existing methods. In models of mammalian trunk development and mouse hematopoiesis, Carta identifies important features of development that are not revealed by other methods including convergent differentiation of specialized cell types, progenitor differentiation dynamics, and the refinement of routes of differentiation via new intermediate progenitors.
Collapse
Affiliation(s)
- Palash Sashittal
- Dept. of Computer Science, Princeton University, Princeton; 08544 NJ, USA
| | - Richard Y. Zhang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton; 08544 NJ, USA
| | - Benjamin K. Law
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton; 08544 NJ, USA
- Dept. of Molecular Biology, Princeton University, Princeton; 08544 NJ, USA
| | | | - Henri Schmidt
- Dept. of Computer Science, Princeton University, Princeton; 08544 NJ, USA
| | - Adriano Bolondi
- Dept. of Genome Regulation, Max Planck Institute for Molecular Genetics; 14195 Berlin, Germany
| | - Michelle M. Chan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton; 08544 NJ, USA
- Dept. of Molecular Biology, Princeton University, Princeton; 08544 NJ, USA
| | | |
Collapse
|
6
|
Garge RK, Lynch V, Fields R, Casadei S, Best S, Stone J, Snyder M, McGann CD, Shendure J, Starita LM, Hamazaki N, Schweppe DK. The proteomic landscape and temporal dynamics of mammalian gastruloid development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.609098. [PMID: 39282277 PMCID: PMC11398484 DOI: 10.1101/2024.09.05.609098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Gastrulation is the highly coordinated process by which the early embryo breaks symmetry, establishes germ layers and a body plan, and sets the stage for organogenesis. As early mammalian development is challenging to study in vivo, stem cell-derived models have emerged as powerful surrogates, e.g. human and mouse gastruloids. However, although single cell RNA-seq (scRNA-seq) and high-resolution imaging have been extensively applied to characterize such in vitro embryo models, a paucity of measurements of protein dynamics and regulation leaves a major gap in our understanding. Here, we sought to address this by applying quantitative proteomics to human and mouse gastruloids at four key stages of their differentiation (naïve ESCs, primed ESCs, early gastruloids, late gastruloids). To the resulting data, we perform network analysis to map the dynamics of expression of macromolecular protein complexes and biochemical pathways, including identifying cooperative proteins that associate with them. With matched RNA-seq and phosphosite data from these same stages, we investigate pathway-, stage- and species-specific aspects of translational and post-translational regulation, e.g. finding peri-gastrulation stages of human and mice to be discordant with respect to the mitochondrial transcriptome vs. proteome, and nominating novel kinase-substrate relationships based on phosphosite dynamics. Finally, we leverage correlated dynamics to identify conserved protein networks centered around congenital disease genes. Altogether, our data (https://gastruloid.brotmanbaty.org/) and analyses showcase the potential of intersecting in vitro embryo models and proteomics to advance our understanding of early mammalian development in ways not possible through transcriptomics alone.
Collapse
Affiliation(s)
- Riddhiman K. Garge
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Valerie Lynch
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Rose Fields
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Silvia Casadei
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Sabrina Best
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Jeremy Stone
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Matthew Snyder
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Chris D. McGann
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, Washington, USA
- Seattle Hub for Synthetic Biology, Seattle, Washington, USA
| | - Lea M. Starita
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
| | - Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
- Seattle Hub for Synthetic Biology, Seattle, Washington, USA
| | - Devin K. Schweppe
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
7
|
Joshi P, Keyvani Chahi A, Liu L, Moreira S, Vujovic A, Hope KJ. RNA binding protein-directed control of leukemic stem cell evolution and function. Hemasphere 2024; 8:e116. [PMID: 39175825 PMCID: PMC11339706 DOI: 10.1002/hem3.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/06/2024] [Accepted: 05/26/2024] [Indexed: 08/24/2024] Open
Abstract
Strict control over hematopoietic stem cell decision making is essential for healthy life-long blood production and underpins the origins of hematopoietic diseases. Acute myeloid leukemia (AML) in particular is a devastating hematopoietic malignancy that arises from the clonal evolution of disease-initiating primitive cells which acquire compounding genetic changes over time and culminate in the generation of leukemic stem cells (LSCs). Understanding the molecular underpinnings of these driver cells throughout their development will be instrumental in the interception of leukemia, the enabling of effective treatment of pre-leukemic conditions, as well as the development of strategies to target frank AML disease. To this point, a number of precancerous myeloid disorders and age-related alterations are proving as instructive models to gain insights into the initiation of LSCs. Here, we explore this myeloid dysregulation at the level of post-transcriptional control, where RNA-binding proteins (RBPs) function as core effectors. Through regulating the interplay of a myriad of RNA metabolic processes, RBPs orchestrate transcript fates to govern gene expression in health and disease. We describe the expanding appreciation of the role of RBPs and their post-transcriptional networks in sustaining healthy hematopoiesis and their dysregulation in the pathogenesis of clonal myeloid disorders and AML, with a particular emphasis on findings described in human stem cells. Lastly, we discuss key breakthroughs that highlight RBPs and post-transcriptional control as actionable targets for precision therapy of AML.
Collapse
Affiliation(s)
- Pratik Joshi
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Ava Keyvani Chahi
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Lina Liu
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Steven Moreira
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Ana Vujovic
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| | - Kristin J. Hope
- Department of Medical BiophysicsUniversity of TorontoTorontoCanada
- Princess Margaret Cancer CenterUniversity Health NetworkTorontoCanada
| |
Collapse
|
8
|
Watanuki S, Kobayashi H, Sugiura Y, Yamamoto M, Karigane D, Shiroshita K, Sorimachi Y, Morikawa T, Fujita S, Shide K, Haraguchi M, Tamaki S, Mikawa T, Kondoh H, Nakano H, Sumiyama K, Nagamatsu G, Goda N, Okamoto S, Nakamura-Ishizu A, Shimoda K, Suematsu M, Suda T, Takubo K. SDHAF1 confers metabolic resilience to aging hematopoietic stem cells by promoting mitochondrial ATP production. Cell Stem Cell 2024; 31:1145-1161.e15. [PMID: 38772377 DOI: 10.1016/j.stem.2024.04.023] [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: 04/27/2023] [Revised: 02/20/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024]
Abstract
Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.
Collapse
Affiliation(s)
- Shintaro Watanuki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka 564-8565, Japan
| | - Daiki Karigane
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kohei Shiroshita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yuriko Sorimachi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and Engineering, Tokyo 162-8480, Japan
| | - Takayuki Morikawa
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shinya Fujita
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kotaro Shide
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Miho Haraguchi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Shinpei Tamaki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan
| | - Takumi Mikawa
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroshi Kondoh
- Geriatric Unit, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Toho University School of Medicine, Tokyo 143-8540, Japan
| | - Kenta Sumiyama
- Laboratory of Animal Genetics and Breeding, Department of Animal Sciences, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan; RIKEN Center for Biosystems Dynamics Research, Laboratory for Mouse Genetic Engineering, Osaka 565-0871, Japan
| | - Go Nagamatsu
- Center for Advanced Assisted Reproductive Technologies, University of Yamanashi, Kofu 400-8501, Japan
| | - Nobuhito Goda
- Department of Life Sciences and Medical BioScience, Waseda University School of Advanced Science and Engineering, Tokyo 162-8480, Japan
| | - Shinichiro Okamoto
- Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ayako Nakamura-Ishizu
- Department of Microscopic and Developmental Anatomy, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Kazuya Shimoda
- Division of Hematology, Diabetes, and Endocrinology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Live Imaging Center, Central Institute for Experimental Medicine and Life Science, Kawasaki 210-0821, Japan
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, Tokyo 162-8655, Japan; Department of Cell Fate Biology and Stem Cell Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan.
| |
Collapse
|
9
|
Gill JS, Bansal B, Guo K, Huang F, Singh H, Hur J, Khan N, Mathur R. Mitochondrial Oxidative Stress Regulates FOXP3+ T-Cell Activity and CD4-Mediated Inflammation in Older Adults with Frailty. Int J Mol Sci 2024; 25:6235. [PMID: 38892421 PMCID: PMC11173216 DOI: 10.3390/ijms25116235] [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: 04/23/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
In healthy older adults, the immune system generally preserves its response and contributes to a long, healthy lifespan. However, rapid deterioration in immune regulation can lead to chronic inflammation, termed inflammaging, which accelerates pathological aging and diminishes the quality of life in older adults with frailty. A significant limitation in current aging research is the predominant focus on comparisons between young and older populations, often overlooking the differences between healthy older adults and those experiencing pathological aging. Our study elucidates the intricate immunological dynamics of the CD4/Treg axis in frail older adults compared to comparable age-matched healthy older adults. By utilizing publicly available RNA sequencing and single-cell RNA sequencing (scRNAseq) data from peripheral blood mononuclear cells (PBMCs), we identified a specific Treg cell subset and transcriptional landscape contributing to the dysregulation of CD4+ T-cell responses. We explored the molecular mechanisms underpinning Treg dysfunction, revealing that Tregs from frail older adults exhibit reduced mitochondrial protein levels, impairing mitochondrial oxidative phosphorylation. This impairment is driven by the TNF/NF-kappa B pathway, leading to cumulative inflammation. Further, we gained a deeper understanding of the CD4/Treg axis by predicting the effects of gene perturbations on cellular signaling networks. Collectively, these findings highlight the age-related relationship between mitochondrial dysfunction in the CD4/Treg axis and its role in accelerating aging and frailty in older adults. Targeting Treg dysfunction offers a critical basis for developing tailored therapeutic strategies aimed at improving the quality of life in older adults.
Collapse
Affiliation(s)
- Jappreet Singh Gill
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
- Department of Biomedical Engineering, School of Electrical Engineering and Computer Sciences, University of North Dakota, Grand Forks, ND 58292, USA
| | - Benu Bansal
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
- Department of Biomedical Engineering, School of Electrical Engineering and Computer Sciences, University of North Dakota, Grand Forks, ND 58292, USA
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Kai Guo
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fang Huang
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Harpreet Singh
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (K.G.); (F.H.); (J.H.)
| | - Nadeem Khan
- Department of Oral Biology, University of Florida, Gainsville, FL 32603, USA;
| | - Ramkumar Mathur
- Department of Geriatrics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (J.S.G.); (B.B.); (H.S.)
| |
Collapse
|
10
|
Ye L, Tian C, Li Y, Pan H, Hu J, Shu L, Pan X. Hematopoietic aging: Cellular, molecular, and related mechanisms. Chin Med J (Engl) 2024; 137:1303-1312. [PMID: 37898877 PMCID: PMC11191024 DOI: 10.1097/cm9.0000000000002871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Indexed: 10/30/2023] Open
Abstract
ABSTRACT Aging is accompanied by significant inhibition of hematopoietic and immune system function and disruption of bone marrow structure. Aging-related alterations in the inflammatory response, immunity, and stem cell niches are at the root of hematopoietic aging. Understanding the molecular mechanisms underlying hematopoietic and bone marrow aging can aid the clinical treatment of aging-related diseases. In particular, it is unknown how the niche reprograms hematopoietic stem cells (HSCs) in an age-dependent manner to maintain normal hematopoiesis in elderly individuals. Recently, specific inhibitors and blood exchange methods have been shown to reshape the hematopoietic niche and reverse hematopoietic aging. Here, we present the latest scientific discoveries related to hematopoietic aging and hematopoietic system rejuvenation, discuss the relationships between hematopoietic niche aging and HSC aging, and describe related studies on stem cell-mediated regulation of hematopoietic aging, aiming to provide new ideas for further study.
Collapse
Affiliation(s)
- Li Ye
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
- Department of Immunology of School of Basic Medicine of Guizhou Medical University, National and Local Joint Engineering Laboratory of Cell Engineering Biomedical Technology, Key Laboratory of Regenerative Medicine of Guizhou Province, State Key Laboratory of Efficacy and Utilization of Medicinal Plants Co-constructed by Province and Ministry, Key Laboratory of Translational Research of Adult Stem Cell of Chinese Academy of Medical Sciences, Guiyang, Guizhou 550025, China
| | - Chuan Tian
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
| | - Ye Li
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
- Department of Immunology of School of Basic Medicine of Guizhou Medical University, National and Local Joint Engineering Laboratory of Cell Engineering Biomedical Technology, Key Laboratory of Regenerative Medicine of Guizhou Province, State Key Laboratory of Efficacy and Utilization of Medicinal Plants Co-constructed by Province and Ministry, Key Laboratory of Translational Research of Adult Stem Cell of Chinese Academy of Medical Sciences, Guiyang, Guizhou 550025, China
| | - Hang Pan
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
- Department of Immunology of School of Basic Medicine of Guizhou Medical University, National and Local Joint Engineering Laboratory of Cell Engineering Biomedical Technology, Key Laboratory of Regenerative Medicine of Guizhou Province, State Key Laboratory of Efficacy and Utilization of Medicinal Plants Co-constructed by Province and Ministry, Key Laboratory of Translational Research of Adult Stem Cell of Chinese Academy of Medical Sciences, Guiyang, Guizhou 550025, China
| | - Jinxiu Hu
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
| | - Liping Shu
- Department of Immunology of School of Basic Medicine of Guizhou Medical University, National and Local Joint Engineering Laboratory of Cell Engineering Biomedical Technology, Key Laboratory of Regenerative Medicine of Guizhou Province, State Key Laboratory of Efficacy and Utilization of Medicinal Plants Co-constructed by Province and Ministry, Key Laboratory of Translational Research of Adult Stem Cell of Chinese Academy of Medical Sciences, Guiyang, Guizhou 550025, China
| | - Xinghua Pan
- The Stem Cells and Immune Cells Biomedical Techniques Integrated Engineering Laboratory of State and Regions, Cell Therapy Technology Transfer Medical Key Laboratory of Yunnan Province, Kunming Key Laboratory of Stem Cell and Regenerative Medicine, Basic Medical Laboratory, 920th Hospital of Joint Logistics Support Force, PLA, Kunming, Yunnan 650032, China
| |
Collapse
|
11
|
Yamada Y, Zheng Z, Jad AK, Yamashita M. Lethal and sublethal effects of programmed cell death pathways on hematopoietic stem cells. Exp Hematol 2024; 134:104214. [PMID: 38582294 DOI: 10.1016/j.exphem.2024.104214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Programmed cell death is an evolutionally conserved cellular process in multicellular organisms that eliminates unnecessary or rogue cells during development, infection, and carcinogenesis. Hematopoietic stem cells (HSCs) are a rare, self-renewing, and multipotent cell population necessary for the establishment and regeneration of the hematopoietic system. Counterintuitively, key components necessary for programmed cell death induction are abundantly expressed in long-lived HSCs, which often survive myeloablative stress by engaging a prosurvival response that counteracts cell death-inducing stimuli. Although HSCs are well known for their apoptosis resistance, recent studies have revealed their unique vulnerability to certain types of programmed necrosis, such as necroptosis and ferroptosis. Moreover, emerging evidence has shown that programmed cell death pathways can be sublethally activated to cause nonlethal consequences such as innate immune response, organelle dysfunction, and mutagenesis. In this review, we summarized recent findings on how divergent cell death programs are molecularly regulated in HSCs. We then discussed potential side effects caused by sublethal activation of programmed cell death pathways on the functionality of surviving HSCs.
Collapse
Affiliation(s)
- Yuta Yamada
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Zhiqian Zheng
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Alaa K Jad
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masayuki Yamashita
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
12
|
Zhou H, Li I, Bramlett CS, Wang B, Hao J, Yen DP, Ando Y, Fraser SE, Lu R, Shen K. Label-free metabolic optical biomarkers track stem cell fate transition in real time. SCIENCE ADVANCES 2024; 10:eadi6770. [PMID: 38718114 PMCID: PMC11078180 DOI: 10.1126/sciadv.adi6770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Tracking stem cell fate transition is crucial for understanding their development and optimizing biomanufacturing. Destructive single-cell methods provide a pseudotemporal landscape of stem cell differentiation but cannot monitor stem cell fate in real time. We established a metabolic optical metric using label-free fluorescence lifetime imaging microscopy (FLIM), feature extraction and machine learning-assisted analysis, for real-time cell fate tracking. From a library of 205 metabolic optical biomarker (MOB) features, we identified 56 associated with hematopoietic stem cell (HSC) differentiation. These features collectively describe HSC fate transition and detect its bifurcate lineage choice. We further derived a MOB score measuring the "metabolic stemness" of single cells and distinguishing their division patterns. This score reveals a distinct role of asymmetric division in rescuing stem cells with compromised metabolic stemness and a unique mechanism of PI3K inhibition in promoting ex vivo HSC maintenance. MOB profiling is a powerful tool for tracking stem cell fate transition and improving their biomanufacturing from a single-cell perspective.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Irene Li
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Charles S. Bramlett
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Bowen Wang
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jia Hao
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Daniel P. Yen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Yuta Ando
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Scott E. Fraser
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089, USA
- Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Rong Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Keyue Shen
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
- USC Stem Cell, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
13
|
Maneix L, Iakova P, Lee CG, Moree SE, Lu X, Datar GK, Hill CT, Spooner E, King JCK, Sykes DB, Saez B, Di Stefano B, Chen X, Krause DS, Sahin E, Tsai FTF, Goodell MA, Berk BC, Scadden DT, Catic A. Cyclophilin A supports translation of intrinsically disordered proteins and affects haematopoietic stem cell ageing. Nat Cell Biol 2024; 26:593-603. [PMID: 38553595 PMCID: PMC11021199 DOI: 10.1038/s41556-024-01387-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: 04/25/2022] [Accepted: 02/23/2024] [Indexed: 04/11/2024]
Abstract
Loss of protein function is a driving force of ageing. We have identified peptidyl-prolyl isomerase A (PPIA or cyclophilin A) as a dominant chaperone in haematopoietic stem and progenitor cells. Depletion of PPIA accelerates stem cell ageing. We found that proteins with intrinsically disordered regions (IDRs) are frequent PPIA substrates. IDRs facilitate interactions with other proteins or nucleic acids and can trigger liquid-liquid phase separation. Over 20% of PPIA substrates are involved in the formation of supramolecular membrane-less organelles. PPIA affects regulators of stress granules (PABPC1), P-bodies (DDX6) and nucleoli (NPM1) to promote phase separation and increase cellular stress resistance. Haematopoietic stem cell ageing is associated with a post-transcriptional decrease in PPIA expression and reduced translation of IDR-rich proteins. Here we link the chaperone PPIA to the synthesis of intrinsically disordered proteins, which indicates that impaired protein interaction networks and macromolecular condensation may be potential determinants of haematopoietic stem cell ageing.
Collapse
Affiliation(s)
- Laure Maneix
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Polina Iakova
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Charles G Lee
- Department of BioSciences, Rice University, Houston, TX, USA
| | - Shannon E Moree
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Xuan Lu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Gandhar K Datar
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Cedric T Hill
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric Spooner
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Jordon C K King
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Borja Saez
- Center for Applied Medical Research, Hematology-Oncology Unit, Pamplona, Navarra, Spain
| | - Bruno Di Stefano
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Ergun Sahin
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Francis T F Tsai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Margaret A Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA
| | - Bradford C Berk
- Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - André Catic
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA.
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Cell and Gene Therapy Program at the Dan L. Duncan Comprehensive Cancer Center, Houston, TX, USA.
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA.
| |
Collapse
|
14
|
Ross JB, Myers LM, Noh JJ, Collins MM, Carmody AB, Messer RJ, Dhuey E, Hasenkrug KJ, Weissman IL. Depleting myeloid-biased haematopoietic stem cells rejuvenates aged immunity. Nature 2024; 628:162-170. [PMID: 38538791 PMCID: PMC11870232 DOI: 10.1038/s41586-024-07238-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/26/2024] [Indexed: 04/01/2024]
Abstract
Ageing of the immune system is characterized by decreased lymphopoiesis and adaptive immunity, and increased inflammation and myeloid pathologies1,2. Age-related changes in populations of self-renewing haematopoietic stem cells (HSCs) are thought to underlie these phenomena3. During youth, HSCs with balanced output of lymphoid and myeloid cells (bal-HSCs) predominate over HSCs with myeloid-biased output (my-HSCs), thereby promoting the lymphopoiesis required for initiating adaptive immune responses, while limiting the production of myeloid cells, which can be pro-inflammatory4. Ageing is associated with increased proportions of my-HSCs, resulting in decreased lymphopoiesis and increased myelopoiesis3,5,6. Transfer of bal-HSCs results in abundant lymphoid and myeloid cells, a stable phenotype that is retained after secondary transfer; my-HSCs also retain their patterns of production after secondary transfer5. The origin and potential interconversion of these two subsets is still unclear. If they are separate subsets postnatally, it might be possible to reverse the ageing phenotype by eliminating my-HSCs in aged mice. Here we demonstrate that antibody-mediated depletion of my-HSCs in aged mice restores characteristic features of a more youthful immune system, including increasing common lymphocyte progenitors, naive T cells and B cells, while decreasing age-related markers of immune decline. Depletion of my-HSCs in aged mice improves primary and secondary adaptive immune responses to viral infection. These findings may have relevance to the understanding and intervention of diseases exacerbated or caused by dominance of the haematopoietic system by my-HSCs.
Collapse
Affiliation(s)
- Jason B Ross
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Lara M Myers
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Joseph J Noh
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madison M Collins
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- Department of Biological and Physical Sciences, Montana State University Billings, Billings, MT, USA
| | - Aaron B Carmody
- Research Technologies Branch, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ronald J Messer
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Erica Dhuey
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Kim J Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
15
|
Tang B, Wang X, He H, Chen R, Qiao G, Yang Y, Xu Z, Wang L, Dong Q, Yu J, Zhang MQ, Shi M, Wang J. Aging-disturbed FUS phase transition impairs hematopoietic stem cells by altering chromatin structure. Blood 2024; 143:124-138. [PMID: 37748139 DOI: 10.1182/blood.2023020539] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/08/2023] [Accepted: 09/09/2023] [Indexed: 09/27/2023] Open
Abstract
ABSTRACT Aged hematopoietic stem cells (HSCs) exhibit compromised reconstitution capacity. The molecular mechanisms behind this phenomenon are not fully understood. Here, we observed that the expression of FUS is increased in aged HSCs, and enforced FUS recapitulates the phenotype of aged HSCs through arginine-glycine-glycine-mediated aberrant FUS phase transition. By using Fus-gfp mice, we observed that FUShigh HSCs exhibit compromised FUS mobility and resemble aged HSCs both functionally and transcriptionally. The percentage of FUShigh HSCs is increased upon physiological aging and replication stress, and FUSlow HSCs of aged mice exhibit youthful function. Mechanistically, FUShigh HSCs exhibit a different global chromatin organization compared with FUSlow HSCs, which is observed in aged HSCs. Many topologically associating domains (TADs) are merged in aged HSCs because of the compromised binding of CCCTC-binding factor with chromatin, which is invoked by aberrant FUS condensates. It is notable that the transcriptional alteration between FUShigh and FUSlow HSCs originates from the merged TADs and is enriched in HSC aging-related genes. Collectively, this study reveals for the first time that aberrant FUS mobility promotes HSC aging by altering chromatin structure.
Collapse
Affiliation(s)
- Baixue Tang
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xinming Wang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hanqing He
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ruiqing Chen
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Guofeng Qiao
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yang Yang
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Zihan Xu
- School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, Peking University, Beijing, China
| | - Longteng Wang
- School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, Peking University, Beijing, China
| | - Qiongye Dong
- Institute of Precision of Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jia Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Michael Q Zhang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, Beijing National Research Center for Information Science and Technology, School of Medicine, Tsinghua University, Beijing, China
- Department of Biological Sciences, Center for Systems Biology, The University of Texas, Richardson, TX
| | - Minglei Shi
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, Beijing National Research Center for Information Science and Technology, School of Medicine, Tsinghua University, Beijing, China
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Jianwei Wang
- Department of Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
16
|
Karimnia N, Harris J, Heazlewood SY, Cao B, Nilsson SK. Metabolic regulation of aged hematopoietic stem cells: key players and mechanisms. Exp Hematol 2023; 128:2-9. [PMID: 37778498 DOI: 10.1016/j.exphem.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Affiliation(s)
- Nazanin Karimnia
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - James Harris
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia; School of Clinical Sciences, Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia
| | - Shen Y Heazlewood
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
| | - Benjamin Cao
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia.
| | - Susan K Nilsson
- Biomedical Manufacturing, Commonwealth Scientific and Industrial Research Organisation, Clayton, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, Australia.
| |
Collapse
|
17
|
Wang Y, Zhang Z, He H, Song J, Cui Y, Chen Y, Zhuang Y, Zhang X, Li M, Zhang X, Zhang MQ, Shi M, Yi C, Wang J. Aging-induced pseudouridine synthase 10 impairs hematopoietic stem cells. Haematologica 2023; 108:2677-2689. [PMID: 37165848 PMCID: PMC10542847 DOI: 10.3324/haematol.2022.282211] [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: 10/04/2022] [Accepted: 05/04/2023] [Indexed: 05/12/2023] Open
Abstract
Aged hematopoietic stem cells (HSC) exhibit compromised reconstitution capacity and differentiation-bias towards myeloid lineage, however, the molecular mechanism behind it remains not fully understood. In this study, we observed that the expression of pseudouridine (Ψ) synthase 10 is increased in aged hematopoietic stem and progenitor cells (HSPC) and enforced protein of Ψ synthase 10 (PUS10) recapitulates the phenotype of aged HSC, which is not achieved by its Ψ synthase activity. Consistently, we observed no difference of transcribed RNA pseudouridylation profile between young and aged HSPC. No significant alteration of hematopoietic homeostasis and HSC function is observed in young Pus10-/- mice, while aged Pus10-/- mice exhibit mild alteration of hematopoietic homeostasis and HSC function. Moreover, we observed that PUS10 is ubiquitinated by E3 ubiquitin ligase CRL4DCAF1 complex and the increase of PUS10 in aged HSPC is due to aging-declined CRL4DCAF1- mediated ubiquitination degradation signaling. Taken together, this study for the first time evaluated the role of PUS10 in HSC aging and function, and provided a novel insight into HSC rejuvenation and its clinical application.
Collapse
Affiliation(s)
- Yuqian Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | | | - Hanqing He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | - Jinghui Song
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093
| | - Yang Cui
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084
| | - Yunan Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing
| | - Yuan Zhuang
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing
| | - Xiaoting Zhang
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing
| | - Mo Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191
| | - Xinxiang Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing
| | - Michael Q Zhang
- School of Medicine, Tsinghua University, Beijing 100084, China; MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist; Department of Automation, Tsinghua University, Beijing 100084, China; Department of Biological Sciences, Center for Systems Biology, the University of Texas, Richardson, TX 75080-3021.
| | - Minglei Shi
- School of Medicine, Tsinghua University, Beijing 100084.
| | - Chengqi Yi
- Department of Basic Medical Sciences, School of Medicine, Institute for Immunology, Beijing Key Lab for Immunological Research on Chronic Diseases, THU-PKU Center for Life Sciences, Tsinghua University, Beijing.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084.
| |
Collapse
|
18
|
Kolabas ZI, Kuemmerle LB, Perneczky R, Förstera B, Ulukaya S, Ali M, Kapoor S, Bartos LM, Büttner M, Caliskan OS, Rong Z, Mai H, Höher L, Jeridi D, Molbay M, Khalin I, Deligiannis IK, Negwer M, Roberts K, Simats A, Carofiglio O, Todorov MI, Horvath I, Ozturk F, Hummel S, Biechele G, Zatcepin A, Unterrainer M, Gnörich J, Roodselaar J, Shrouder J, Khosravani P, Tast B, Richter L, Díaz-Marugán L, Kaltenecker D, Lux L, Chen Y, Zhao S, Rauchmann BS, Sterr M, Kunze I, Stanic K, Kan VWY, Besson-Girard S, Katzdobler S, Palleis C, Schädler J, Paetzold JC, Liebscher S, Hauser AE, Gokce O, Lickert H, Steinke H, Benakis C, Braun C, Martinez-Jimenez CP, Buerger K, Albert NL, Höglinger G, Levin J, Haass C, Kopczak A, Dichgans M, Havla J, Kümpfel T, Kerschensteiner M, Schifferer M, Simons M, Liesz A, Krahmer N, Bayraktar OA, Franzmeier N, Plesnila N, Erener S, Puelles VG, Delbridge C, Bhatia HS, Hellal F, Elsner M, Bechmann I, Ondruschka B, Brendel M, Theis FJ, Erturk A. Distinct molecular profiles of skull bone marrow in health and neurological disorders. Cell 2023; 186:3706-3725.e29. [PMID: 37562402 PMCID: PMC10443631 DOI: 10.1016/j.cell.2023.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/24/2023] [Accepted: 07/07/2023] [Indexed: 08/12/2023]
Abstract
The bone marrow in the skull is important for shaping immune responses in the brain and meninges, but its molecular makeup among bones and relevance in human diseases remain unclear. Here, we show that the mouse skull has the most distinct transcriptomic profile compared with other bones in states of health and injury, characterized by a late-stage neutrophil phenotype. In humans, proteome analysis reveals that the skull marrow is the most distinct, with differentially expressed neutrophil-related pathways and a unique synaptic protein signature. 3D imaging demonstrates the structural and cellular details of human skull-meninges connections (SMCs) compared with veins. Last, using translocator protein positron emission tomography (TSPO-PET) imaging, we show that the skull bone marrow reflects inflammatory brain responses with a disease-specific spatial distribution in patients with various neurological disorders. The unique molecular profile and anatomical and functional connections of the skull show its potential as a site for diagnosing, monitoring, and treating brain diseases.
Collapse
Affiliation(s)
- Zeynep Ilgin Kolabas
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Louis B Kuemmerle
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Robert Perneczky
- Division of Mental Health in Older Adults and Alzheimer Therapy and Research Center, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University Munich, 80336 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Benjamin Förstera
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Selin Ulukaya
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Mayar Ali
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Saketh Kapoor
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Laura M Bartos
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maren Büttner
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Ozum Sehnaz Caliskan
- Institute for Diabetes and Obesity, Helmholtz Center Munich and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Zhouyi Rong
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Medical Research School (MMRS), 80336 Munich, Germany
| | - Hongcheng Mai
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Medical Research School (MMRS), 80336 Munich, Germany
| | - Luciano Höher
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Denise Jeridi
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Muge Molbay
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | | | - Moritz Negwer
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | | | - Alba Simats
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Olga Carofiglio
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Mihail I Todorov
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Izabela Horvath
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; School of Computation, Information and Technology (CIT), TUM, Boltzmannstr. 3, 85748 Garching, Germany
| | - Furkan Ozturk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Selina Hummel
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gloria Biechele
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Artem Zatcepin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jay Roodselaar
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Joshua Shrouder
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Pardis Khosravani
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Benjamin Tast
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lisa Richter
- Biomedical Center (BMC), Core Facility Flow Cytometry, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Laura Díaz-Marugán
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Doris Kaltenecker
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Diabetes and Cancer, Helmholtz Munich, Munich, Germany
| | - Laurin Lux
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Ying Chen
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Shan Zhao
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Boris-Stephan Rauchmann
- Division of Mental Health in Older Adults and Alzheimer Therapy and Research Center, Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University Munich, 80336 Munich, Germany; Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK; Institute of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ines Kunze
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Karen Stanic
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Vanessa W Y Kan
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Sabrina Katzdobler
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Schädler
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes C Paetzold
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Department of Computing, Imperial College London, London, UK
| | - Sabine Liebscher
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Anja E Hauser
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Heiko Lickert
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Stem Cell Research, Helmholtz Zentrum München, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Hanno Steinke
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany
| | - Corinne Benakis
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Christian Braun
- Institute of Legal Medicine, Faculty of Medicine, LMU Munich, Germany
| | - Celia P Martinez-Jimenez
- Helmholtz Pioneer Campus (HPC), Helmholtz Munich, Neuherberg, Germany; TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Neurology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anna Kopczak
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joachim Havla
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Tania Kümpfel
- Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Martin Kerschensteiner
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Institute of Clinical Neuroimmunology, University Hospital Munich, Ludwig-Maximilians University Munich, Munich, Germany; Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians Universität Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Natalie Krahmer
- Institute for Diabetes and Obesity, Helmholtz Center Munich and German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | | | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Suheda Erener
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Claire Delbridge
- Institute of Pathology, Department of Neuropathology, Technical University Munich, TUM School of Medicine, Munich, Germany
| | - Harsharan Singh Bhatia
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Farida Hellal
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Elsner
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, 04109 Leipzig, Germany
| | - Benjamin Ondruschka
- Institute of Legal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Department of Mathematics, Technische Universität München, Garching bei München, Germany
| | - Ali Erturk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Center, Neuherberg, Munich, Germany; Institute for Stroke and Dementia Research, LMU University Hospital, Ludwig-Maximilians University Munich, Munich, Germany; Graduate School of Systemic Neurosciences (GSN), Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
19
|
Colom Díaz PA, Mistry JJ, Trowbridge JJ. Hematopoietic stem cell aging and leukemia transformation. Blood 2023; 142:533-542. [PMID: 36800569 PMCID: PMC10447482 DOI: 10.1182/blood.2022017933] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
With aging, hematopoietic stem cells (HSCs) have an impaired ability to regenerate, differentiate, and produce an entire repertoire of mature blood and immune cells. Owing to dysfunctional hematopoiesis, the incidence of hematologic malignancies increases among elderly individuals. Here, we provide an update on HSC-intrinsic and -extrinsic factors and processes that were recently discovered to contribute to the functional decline of HSCs during aging. In addition, we discuss the targets and timing of intervention approaches to maintain HSC function during aging and the extent to which these same targets may prevent or delay transformation to hematologic malignancies.
Collapse
|
20
|
Mehlferber MM, Kuyumcu-Martinez M, Miller CL, Sheynkman GM. Transcription factors and splice factors - interconnected regulators of stem cell differentiation. CURRENT STEM CELL REPORTS 2023; 9:31-41. [PMID: 38939410 PMCID: PMC11210451 DOI: 10.1007/s40778-023-00227-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 06/29/2024]
Abstract
Purpose of review The underlying molecular mechanisms that direct stem cell differentiation into fully functional, mature cells remain an area of ongoing investigation. Cell state is the product of the combinatorial effect of individual factors operating within a coordinated regulatory network. Here, we discuss the contribution of both gene regulatory and splicing regulatory networks in defining stem cell fate during differentiation and the critical role of protein isoforms in this process. Recent findings We review recent experimental and computational approaches that characterize gene regulatory networks, splice regulatory networks, and the resulting transcriptome and proteome they mediate during differentiation. Such approaches include long-read RNA sequencing, which has demonstrated high-resolution profiling of mRNA isoforms, and Cas13-based CRISPR, which could make possible high-throughput isoform screening. Collectively, these developments enable systems-level profiling of factors contributing to cell state. Summary Overall, gene and splice regulatory networks are important in defining cell state. The emerging high-throughput systems-level approaches will characterize the gene regulatory network components necessary in driving stem cell differentiation.
Collapse
Affiliation(s)
- Madison M Mehlferber
- Department of Biochemistry and Molecular Genetics, University Virginia, Charlottesville, VA 22903
| | - Muge Kuyumcu-Martinez
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Fontaine Medical Office Building 1, 415 Ray C. Hunt Dr, Charlottesville, VA 22903
| | - Clint L Miller
- Department of Public Health Sciences, Department of Biochemistry and Molecular Genetics, and Department of Biomedical Engineering, University of Virginia, Multistory Building, West Complex, 1335 Lee St, Charlottesville, VA 22908, PO Box 800717, Charlottesville, Virginia 22908
| | - Gloria M Sheynkman
- Department of Molecular Physiology and Biological Physics, Center for Public Health Genomics, UVA Comprehensive Cancer Center, Department of Biochemistry and Molecular Genetics, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, VA 22903
| |
Collapse
|
21
|
Bonazzi S, d'Hennezel E, Beckwith REJ, Xu L, Fazal A, Magracheva A, Ramesh R, Cernijenko A, Antonakos B, Bhang HEC, Caro RG, Cobb JS, Ornelas E, Ma X, Wartchow CA, Clifton MC, Forseth RR, Fortnam BH, Lu H, Csibi A, Tullai J, Carbonneau S, Thomsen NM, Larrow J, Chie-Leon B, Hainzl D, Gu Y, Lu D, Meyer MJ, Alexander D, Kinyamu-Akunda J, Sabatos-Peyton CA, Dales NA, Zécri FJ, Jain RK, Shulok J, Wang YK, Briner K, Porter JA, Tallarico JA, Engelman JA, Dranoff G, Bradner JE, Visser M, Solomon JM. Discovery and characterization of a selective IKZF2 glue degrader for cancer immunotherapy. Cell Chem Biol 2023; 30:235-247.e12. [PMID: 36863346 DOI: 10.1016/j.chembiol.2023.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 12/15/2022] [Accepted: 02/09/2023] [Indexed: 03/04/2023]
Abstract
Malignant tumors can evade destruction by the immune system by attracting immune-suppressive regulatory T cells (Treg) cells. The IKZF2 (Helios) transcription factor plays a crucial role in maintaining function and stability of Treg cells, and IKZF2 deficiency reduces tumor growth in mice. Here we report the discovery of NVP-DKY709, a selective molecular glue degrader of IKZF2 that spares IKZF1/3. We describe the recruitment-guided medicinal chemistry campaign leading to NVP-DKY709 that redirected the degradation selectivity of cereblon (CRBN) binders from IKZF1 toward IKZF2. Selectivity of NVP-DKY709 for IKZF2 was rationalized by analyzing the DDB1:CRBN:NVP-DKY709:IKZF2(ZF2 or ZF2-3) ternary complex X-ray structures. Exposure to NVP-DKY709 reduced the suppressive activity of human Treg cells and rescued cytokine production in exhausted T-effector cells. In vivo, treatment with NVP-DKY709 delayed tumor growth in mice with a humanized immune system and enhanced immunization responses in cynomolgus monkeys. NVP-DKY709 is being investigated in the clinic as an immune-enhancing agent for cancer immunotherapy.
Collapse
Affiliation(s)
- Simone Bonazzi
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA.
| | - Eva d'Hennezel
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA.
| | | | - Lei Xu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Aleem Fazal
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Anna Magracheva
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Radha Ramesh
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | - Hyo-Eun C Bhang
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Jennifer S Cobb
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Xiaolei Ma
- Novartis Institutes for Biomedical Research, Emeryville, CA, USA
| | | | | | - Ry R Forseth
- Novartis Institutes for Biomedical Research, East Hanover, NJ, USA
| | | | - Hongbo Lu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Alfredo Csibi
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Jennifer Tullai
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Seth Carbonneau
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Noel M Thomsen
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Jay Larrow
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Dominik Hainzl
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Yi Gu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Darlene Lu
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Matthew J Meyer
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Dylan Alexander
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | - Natalie A Dales
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | - Rishi K Jain
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Janine Shulok
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Y Karen Wang
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Karin Briner
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Glenn Dranoff
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - James E Bradner
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | - Michael Visser
- Novartis Institutes for Biomedical Research, Cambridge, MA, USA
| | | |
Collapse
|
22
|
Grey W, Atkinson S, Rix B, Casado P, Ariza-McNaughton L, Hawley C, Sopoena ML, Bridge KS, Kent D, Cutillas PR, Bonnet D. The CKS1/CKS2 Proteostasis Axis Is Crucial to Maintain Hematopoietic Stem Cell Function. Hemasphere 2023; 7:e853. [PMID: 36874381 PMCID: PMC9977483 DOI: 10.1097/hs9.0000000000000853] [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: 09/12/2022] [Accepted: 01/30/2023] [Indexed: 03/04/2023] Open
Abstract
Long-term hematopoietic stem cells are rare, highly quiescent stem cells of the hematopoietic system with life-long self-renewal potential and the ability to transplant and reconstitute the entire hematopoietic system of conditioned recipients. Most of our understanding of these rare cells has relied on cell surface identification, epigenetic, and transcriptomic analyses. Our knowledge of protein synthesis, folding, modification, and degradation-broadly termed protein homeostasis or "proteostasis"-in these cells is still in its infancy, with very little known about how the functional state of the proteome is maintained in hematopoietic stem cells. We investigated the requirement of the small phospho-binding adaptor proteins, the cyclin-dependent kinase subunits (CKS1 and CKS2), for maintaining ordered hematopoiesis and long-term hematopoietic stem cell reconstitution. CKS1 and CKS2 are best known for their roles in p27 degradation and cell cycle regulation, and by studying the transcriptome and proteome of Cks1 -/- and Cks2 -/- mice, we demonstrate regulation of key signaling pathways that govern hematopoietic stem cell biology including AKT, FOXO1, and NFκB, together balancing protein homeostasis and restraining reactive oxygen species to ensure healthy hematopoietic stem cell function.
Collapse
Affiliation(s)
- William Grey
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Samantha Atkinson
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Beatrice Rix
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro Casado
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | | | - Cathy Hawley
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Miriam L. Sopoena
- Bioinformatics Core, The Francis Crick Institute, London, United Kingdom
| | - Katherine S. Bridge
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - David Kent
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro R. Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | - Dominique Bonnet
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
23
|
Wittorf KJ, Weber KK, Swenson SA, Buckley SM. Ubiquitin E3 ligase FBXO21 regulates cytokine-mediated signaling pathways, but is dispensable for steady-state hematopoiesis. Exp Hematol 2022; 114:33-42.e3. [PMID: 35987460 DOI: 10.1016/j.exphem.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/24/2022]
Abstract
Hematopoietic cell fate decisions such as self-renewal and differentiation are highly regulated through multiple molecular pathways. One pathway, the ubiquitin proteasome system (UPS), controls protein levels by tagging them with polyubiquitin chains and promoting their degradation through the proteasome. Ubiquitin E3 ligases serve as the substrate-recognition component of the UPS. By investigating the FBOX family of E3 ligases, we discovered that Fbxo21 was highly expressed in the hematopoietic stem and progenitor cell (HSPC) population, and exhibited low to no expression in mature myeloid populations. To determine the role of FBXO21 on HSPC maintenance, self-renewal, and differentiation, we generated shRNAs against FBXO21 and a hematopoiesis-specific Fbxo21 conditional knockout (cKO) mouse model. We found that silencing FBXO21 in HSPCs led to a loss in colony formation and an increase in cell differentiation in vitro. Additionally, stressing the HSPC populations in our Fbxo21 cKO mouse with 5-fluorouracil injections resulted in a decrease in survival, despite these populations exhibiting minimal alterations during steady-state hematopoiesis. Although FBXO21 has previously been proposed to regulate cytokine signaling via ASK and p38, our results indicate that depletion of FBXO21 led to altered ERK signaling in vitro. Together, these findings suggest ubiquitin E3 ligase FBXO21 regulates HSPCs through cytokine-mediated pathways.
Collapse
Affiliation(s)
- Karli J Wittorf
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Kasidy K Weber
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Samantha A Swenson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE
| | - Shannon M Buckley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE.
| |
Collapse
|
24
|
Refining the migration and engraftment of short-term and long-term HSCs by enhancing homing-specific adhesion mechanisms. Blood Adv 2022; 6:4373-4391. [PMID: 35764498 DOI: 10.1182/bloodadvances.2022007465] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
In contrast to the short-term(ST)-CD34pos stem cells, studies have suggested that long-term (LT) hematopoietic stem cells (HSC) found in the CD34neg stem cell pool have trouble migrating and engrafting when introduced intravenously. We set out to fully elucidate the adhesion mechanisms used by ST/LT-HSCs to migrate to the bone marrow in order to understand these deficiencies. Focusing on murine ST-HSCs(Flk2negCD34pos) and LT-HSCs(Flk2negCD34neg), we observed a distinctive expression pattern of bone marrow homing effectors necessary for the first step, namely sialyl Lewis-X(sLex;ligand for E-selectin), and the second step, namely CXCR4 (receptor for SDF-1). sLex expression was higher on Flk2negCD34pos ST-HSCs(>60%) compared to Flk2negCD34neg LT-HSCs(<10%), which correlated to binding to E-selectin. Higher levels of CXCR4 were observed on Flk2negCD34pos ST-HSCs compared to Flk2negCD34neg LT-HSCs. Interestingly, expression of CD26, a peptidase known to deactivate chemokines (i.e.SDF-1), was higher on Flk2negCD34neg LT-HSCs. Given that migration is compromised in Flk2negCD34neg LT-HSCs, we aimed to enhance their ability to migrate using recombinant fucosyltransferase 6 (rhFTVI) and DiprotinA (CD26-inhibitor). We observed that although LT-HSCs expressed low levels of sLex, in vivo engraftment was not compromised. Moreover, although both treaments enhanced migration in vitro, only pre-treatment of LT-HSCs with DiprotinA enhanced engraftment in vivo. Remarkably, fucosylation of Flk2negCD34pos ST-HSCs consistently led to their ability to transplant secondary recipients, the gold standard for testing functionality of LT-HSCs. These data suggest that treatments to overcome the molecular disparity in adhesion mechanisms among ST-HSCs and LT-HSCs, differentially influences their abilities to migrate and engraft in vivo and boosts ST-HSCs engraftment in vivo.
Collapse
|
25
|
Volk RF, Montaño JL, Warrington SE, Hofmann KL, Zaro BW. Proteomic characterization of phagocytic primary human monocyte-derived macrophages. RSC Chem Biol 2022; 3:783-793. [PMID: 35755185 PMCID: PMC9175098 DOI: 10.1039/d2cb00076h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/12/2022] [Indexed: 11/21/2022] Open
Abstract
Macrophages play a vital role in the innate immune system, identifying and destroying unwanted cells. However, it has been difficult to attain a comprehensive understanding of macrophage protein abundance due to technical limitations. In addition, it remains unclear how changes in proteome composition are linked to phagocytic activity. In this study we developed methods to derive human macrophages and prepare them for mass spectrometry analysis in order to more-deeply understand the proteomic consequences of macrophage stimulation. Interferon gamma (IF-g), an immune stimulating cytokine, was used to induce macrophage activation, increasing phagocytosis of cancer cells by 2-fold. These conditions were used to perform comparative shotgun proteomics between resting macrophages and stimulated macrophages with increased phagocytic activity. Our analysis revealed that macrophages bias their protein production toward biological processes associated with phagocytosis and antigen processing in response to stimulation. We confirmed our findings by antibody-based western blotting experiments, validating both previously reported and novel proteins of interest. In addition to whole protein changes, we evaluated active protein synthesis by treating cells with the methionine surrogate probe homopropargylglycine (HPG). We saw increased rates of HPG incorporation during phagocytosis-inducing stimulation, suggesting protein synthesis rates are altered by stimulation. Together our findings provide the most comprehensive proteomic insight to date into primary human macrophages. We anticipate that this data can be used as a launchpoint to generate new hypotheses about innate immune function.
Collapse
Affiliation(s)
- Regan F Volk
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - José L Montaño
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Sara E Warrington
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Katherine L Hofmann
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| |
Collapse
|
26
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
27
|
Mitochondrial oxidative phosphorylation is dispensable for survival of CD34+ chronic myeloid leukemia stem and progenitor cells. Cell Death Dis 2022; 13:384. [PMID: 35444236 PMCID: PMC9021200 DOI: 10.1038/s41419-022-04842-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 12/24/2022]
Abstract
AbstractChronic myeloid leukemia (CML) are initiated and sustained by self-renewing malignant CD34+ stem cells. Extensive efforts have been made to reveal the metabolic signature of the leukemia stem/progenitor cells in genomic, transcriptomic, and metabolomic studies. However, very little proteomic investigation has been conducted and the mechanism regarding at what level the metabolic program was rewired remains poorly understood. Here, using label-free quantitative proteomic profiling, we compared the signature of CD34+ stem/progenitor cells collected from CML individuals with that of healthy donors and observed significant changes in the abundance of enzymes associated with aerobic central carbonate metabolic pathways. Specifically, CML stem/progenitor cells expressed increased tricarboxylic acid cycle (TCA) with decreased glycolytic proteins, accompanying by increased oxidative phosphorylation (OXPHOS) and decreased glycolysis activity. Administration of the well-known OXPHOS inhibitor metformin eradicated CML stem/progenitor cells and re-sensitized CD34+ CML cells to imatinib in vitro and in patient-derived tumor xenograft murine model. However, different from normal CD34+ cells, the abundance and activity of OXPHOS protein were both unexpectedly elevated with endoplasmic reticulum stress induced by metformin in CML CD34+ cells. The four major aberrantly expressed protein sets, in contrast, were downregulated by metformin in CML CD34+ cells. These data challenged the dependency of OXPHOS for CML CD34+ cell survival and underlined the novel mechanism of metformin. More importantly, it suggested a strong rationale for the use of tyrosine kinase inhibitors in combination with metformin in treating CML.
Collapse
|
28
|
Jassinskaja M, Hansson J. The Opportunity of Proteomics to Advance the Understanding of Intra- and Extracellular Regulation of Malignant Hematopoiesis. Front Cell Dev Biol 2022; 10:824098. [PMID: 35350382 PMCID: PMC8957922 DOI: 10.3389/fcell.2022.824098] [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: 11/28/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Fetal and adult hematopoiesis are regulated by largely distinct sets of cell-intrinsic gene regulatory networks as well as extracellular cues in their respective microenvironment. These ontogeny-specific programs drive hematopoietic stem and progenitor cells (HSPCs) in fetus and adult to divergent susceptibility to initiation and progression of hematological malignancies, such as leukemia. Elucidating how leukemogenic hits disturb the intra- and extracellular programs in HSPCs along ontogeny will provide a better understanding of the causes for age-associated differences in malignant hematopoiesis and facilitate the improvement of strategies for prevention and treatment of pediatric and adult acute leukemia. Here, we review current knowledge of the intrinsic and extrinsic programs regulating normal and malignant hematopoiesis, with a particular focus on the differences between infant and adult acute leukemia. We discuss the recent advances in mass spectrometry-based proteomics and its opportunity for resolving the interplay of cell-intrinsic and niche-associated factors in regulating malignant hematopoiesis.
Collapse
Affiliation(s)
- Maria Jassinskaja
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, Lund, Sweden.,York Biomedical Research Institute, Department of Biology, University of York, York, United Kingdom
| | - Jenny Hansson
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, Lund, Sweden
| |
Collapse
|
29
|
Ma Y, Will B. To Degrade or Not to Degrade DNMT3A. Cancer Discov 2022; 12:23-25. [PMID: 35022208 DOI: 10.1158/2159-8290.cd-21-1430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aberrant DNA cytosine methylation is a critical contributor to compromised tissue regeneration and malignant transformation, particularly during aging. In this issue of Cancer Discovery, Huang and colleagues define a new class of disease-associated DNA (cytosine-5-)-methyltransferase 3 alpha (DNMT3A) variants with decreased de novo DNA methylation activity due increased proteasomal degradation that are able to drive clonal expansion of hematopoietic stem cells.See related article by Huang et al., p. 220.
Collapse
Affiliation(s)
- Yuhong Ma
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York. .,Department of Medicine (Oncology), Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, New York
| |
Collapse
|
30
|
Insufficiency of FZR1 disturbs HSC quiescence by inhibiting ubiquitin-dependent degradation of RUNX1 in aplastic anemia. Leukemia 2021; 36:834-846. [PMID: 34635784 DOI: 10.1038/s41375-021-01445-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 11/08/2022]
Abstract
FZR1 has been implicated as a master regulator of the cell cycle and quiescence, but its roles and molecular mechanisms in the pathogenesis of severe aplastic anemia (SAA) are unclear. Here, we report that FZR1 is downregulated in SAA HSCs compared with healthy control and is associated with decreased quiescence of HSC. Haploinsufficiency of Fzr1 shows impaired quiescence and self-renewal ability of HSC in two Fzr1 heterozygous knockout mouse models. Mechanistically, FZR1 insufficiency inhibits the ubiquitination of RUNX1 protein at lysine 125, leading to the accumulation of RUNX1 protein, which disturbs the quiescence of HSCs in SAA patients. Moreover, downregulation of Runx1 reversed the loss of quiescence and impaired long-term self-renew ability in Fzr1+/- HSCs in vivo and impaired repopulation capacity in BM from SAA patients in vitro. Our findings, therefore, raise the possibility of a decisive role of the FZR1-RUNX1 pathway in the pathogenesis of SAA via deregulation of HSC quiescence.
Collapse
|
31
|
Meier F, Park MA, Mann M. Trapped Ion Mobility Spectrometry and Parallel Accumulation-Serial Fragmentation in Proteomics. Mol Cell Proteomics 2021; 20:100138. [PMID: 34416385 PMCID: PMC8453224 DOI: 10.1016/j.mcpro.2021.100138] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/05/2021] [Accepted: 08/11/2021] [Indexed: 12/14/2022] Open
Abstract
Recent advances in efficiency and ease of implementation have rekindled interest in ion mobility spectrometry, a technique that separates gas phase ions by their size and shape and that can be hybridized with conventional LC and MS. Here, we review the recent development of trapped ion mobility spectrometry (TIMS) coupled to TOF mass analysis. In particular, the parallel accumulation-serial fragmentation (PASEF) operation mode offers unique advantages in terms of sequencing speed and sensitivity. Its defining feature is that it synchronizes the release of ions from the TIMS device with the downstream selection of precursors for fragmentation in a TIMS quadrupole TOF configuration. As ions are compressed into narrow ion mobility peaks, the number of peptide fragment ion spectra obtained in data-dependent or targeted analyses can be increased by an order of magnitude without compromising sensitivity. Taking advantage of the correlation between ion mobility and mass, the PASEF principle also multiplies the efficiency of data-independent acquisition. This makes the technology well suited for rapid proteome profiling, an increasingly important attribute in clinical proteomics, as well as for ultrasensitive measurements down to single cells. The speed and accuracy of TIMS and PASEF also enable precise measurements of collisional cross section values at the scale of more than a million data points and the development of neural networks capable of predicting them based only on peptide sequences. Peptide collisional cross section values can differ for isobaric sequences or positional isomers of post-translational modifications. This additional information may be leveraged in real time to direct data acquisition or in postprocessing to increase confidence in peptide identifications. These developments make TIMS quadrupole TOF PASEF a powerful and expandable platform for proteomics and beyond.
Collapse
Affiliation(s)
- Florian Meier
- Department Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany; Functional Proteomics, Jena University Hospital, Jena, Germany.
| | - Melvin A Park
- Bruker Daltonics Inc, Billerica, Massachusetts, USA.
| | - Matthias Mann
- Department Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Erythropoiesis is a hierarchical process by which hematopoietic stem cells give rise to red blood cells through gradual cell fate restriction and maturation. Deciphering this process requires the establishment of dynamic gene regulatory networks (GRNs) that predict the response of hematopoietic cells to signals from the environment. Although GRNs have historically been derived from transcriptomic data, recent proteomic studies have revealed a major role for posttranscriptional mechanisms in regulating gene expression during erythropoiesis. These new findings highlight the need to integrate proteomic data into GRNs for a refined understanding of erythropoiesis. RECENT FINDINGS Here, we review recent proteomic studies that have furthered our understanding of erythropoiesis with a focus on quantitative mass spectrometry approaches to measure the abundance of transcription factors and cofactors during differentiation. Furthermore, we highlight challenges that remain in integrating transcriptomic, proteomic, and other omics data into a predictive model of erythropoiesis, and discuss the future prospect of single-cell proteomics. SUMMARY Recent proteomic studies have considerably expanded our knowledge of erythropoiesis beyond the traditional transcriptomic-centric perspective. These findings have both opened up new avenues of research to increase our understanding of erythroid differentiation, while at the same time presenting new challenges in integrating multiple layers of information into a comprehensive gene regulatory model.
Collapse
Affiliation(s)
- Marjorie Brand
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Ottawa, ON K1H8L6, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1H8L6, Canada
| | | |
Collapse
|
33
|
Jassinskaja M, Pimková K, Arh N, Johansson E, Davoudi M, Pereira CF, Sitnicka E, Hansson J. Ontogenic shifts in cellular fate are linked to proteotype changes in lineage-biased hematopoietic progenitor cells. Cell Rep 2021; 34:108894. [PMID: 33761361 DOI: 10.1016/j.celrep.2021.108894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/16/2020] [Accepted: 03/02/2021] [Indexed: 12/16/2022] Open
Abstract
The process of hematopoiesis is subject to substantial ontogenic remodeling that is accompanied by alterations in cellular fate during both development and disease. We combine state-of-the-art mass spectrometry with extensive functional assays to gain insight into ontogeny-specific proteomic mechanisms regulating hematopoiesis. Through deep coverage of the cellular proteome of fetal and adult lympho-myeloid multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs), and granulocyte-monocyte progenitors (GMPs), we establish that features traditionally attributed to adult hematopoiesis are conserved across lymphoid and myeloid lineages, whereas generic fetal features are suppressed in GMPs. We reveal molecular and functional evidence for a diminished granulocyte differentiation capacity in fetal LMPPs and GMPs relative to their adult counterparts. Our data indicate an ontogeny-specific requirement of myosin activity for myelopoiesis in LMPPs. Finally, we uncover an ontogenic shift in the monocytic differentiation capacity of GMPs, partially driven by a differential expression of Irf8 during fetal and adult life.
Collapse
Affiliation(s)
- Maria Jassinskaja
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Kristýna Pimková
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Nejc Arh
- Lund Stem Cell Center, Division of Molecular Medicine and Gene Therapy, Lund University, 221 84 Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Emil Johansson
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden; Department of Laboratory Medicine, Lund University, 221 84 Lund, Sweden
| | - Mina Davoudi
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Carlos-Filipe Pereira
- Lund Stem Cell Center, Division of Molecular Medicine and Gene Therapy, Lund University, 221 84 Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, 221 84 Lund, Sweden
| | - Ewa Sitnicka
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden
| | - Jenny Hansson
- Lund Stem Cell Center, Division of Molecular Hematology, Lund University, 221 84 Lund, Sweden.
| |
Collapse
|